1
|
Pitiriga VC, Papamentzelopoulou M, Nikoloudis D, Saldari C, Konstantinakou KE, Vasileiou IV, Tsakris A. Evaluating SARS-CoV-2 T Cell Immunity in COVID-19-Naive Vaccinated Individuals with and Without Spike Protein IgG Antibodies. Pathogens 2025; 14:415. [PMID: 40430736 PMCID: PMC12113767 DOI: 10.3390/pathogens14050415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2025] [Revised: 04/19/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
Background: The effective management of vaccination schedules requires thorough knowledge of an individual's immunoprotection level, including the interaction and persistence of immune responses at both the humoral and cellular levels following SARS-CoV-2 vaccination. This study aimed to investigate the potential relationship between the levels and duration of the SARS-CoV-2 T cell response and IgG measurements in a cohort of COVID-19-naive individuals who had received the SARS-CoV-2 vaccine. Methods: We performed a retrospective descriptive analysis utilizing data retrieved from the electronic medical records of consecutive COVID-19-naive and vaccinated adult individuals who underwent COVID-19 immunity screening at a private healthcare center from September 2021 to September 2022. T cell response was evaluated using the IGRA methodology T-SPOT®.COVID (Oxford Immunotec, Abingdon, Oxfordshire, UK). Results: A retrospective analysis was conducted on a cohort of 262 individuals, comprising 148 females (56.5%) and 114 males (43.5%), with ages ranging from 17 to 92 years (mean age: 59.47 ± 15.5 years). Among the participants, 216/262 (82.4%) tested negative for SARS-CoV-2 IgG antibodies (group A), while 46/262 (17.6%) tested positive (group B). No significant difference was observed between the two groups in the time period post vaccination, with the mean times after vaccination being 136.38 ± 78.68 days in group A and 140.6 ± 79.5 days in group B (T-test, p = 0.74). Among the two groups, a positive T cell reaction against the S antigen was reported in 132/216 (61.1%) participants in group A and 39/46 (84.8%) in group B (X2 test, p = 0.002). Additionally, individuals with a positive antibody response demonstrated statistically significant higher T SPOT results compared to those with undetectable antibody levels, with a mean SFC count of 125.70 for group A and 158.73 for group B (Mann-Whitney test, p = 0.006). Conclusions: Our findings suggest that T cell immunity may persist even when antibodies are undetectable, highlighting the potential role of cellular immunity in providing protection against COVID-19 over time. Additionally, this study demonstrates a significant correlation between humoral and cellular immune response levels to SARS-CoV-2, suggesting that the activation of humoral immunity following SARS-CoV-2 vaccination is associated with higher levels of cellular immunity compared to individuals with undetectable antibody levels.
Collapse
Affiliation(s)
- Vassiliki C. Pitiriga
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece; (C.S.); (A.T.)
| | - Myrto Papamentzelopoulou
- Molecular Biology Unit, 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Dimitris Nikoloudis
- Bioiatriki Healthcare Group, Kifisias 132 and Papada Street, 11526 Athens, Greece; (D.N.); (K.E.K.); (I.V.V.)
| | - Chrysa Saldari
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece; (C.S.); (A.T.)
| | - Kanella E. Konstantinakou
- Bioiatriki Healthcare Group, Kifisias 132 and Papada Street, 11526 Athens, Greece; (D.N.); (K.E.K.); (I.V.V.)
| | - Irene V. Vasileiou
- Bioiatriki Healthcare Group, Kifisias 132 and Papada Street, 11526 Athens, Greece; (D.N.); (K.E.K.); (I.V.V.)
| | - Athanasios Tsakris
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece; (C.S.); (A.T.)
| |
Collapse
|
2
|
Zhang Y, He Y, Jiao Y, Yang G, Pu Y, Wan Z, Li S, Wu Y, Liao W, Guo J. Surface immobilization of single atoms on heteroatom-doped carbon nanospheres through phenolic-mediated interfacial anchoring for highly efficient biocatalysis. Chem Sci 2025; 16:3479-3489. [PMID: 39877823 PMCID: PMC11770272 DOI: 10.1039/d4sc07775j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/17/2025] [Indexed: 01/31/2025] Open
Abstract
Single-atom catalysts (SACs) dispersed on support materials exhibit exceptional catalytic properties that can be fine-tuned through interactions between the single atoms and the support. However, selectively controlling the spatial location of single metal atoms while simultaneously harmonizing their coordination environment remains a significant challenge. Here, we present a phenolic-mediated interfacial anchoring (PIA) strategy to prepare SACs with Fe single atoms anchored on the surface of heteroatom-doped carbon nanospheres. Briefly, by exploiting metal-phenolic networks (MPNs) for surface coating and phloroglucinol-induced polymerization for support precursor formation, we successfully anchored Fe single atoms at the interface between the MPN layer and the support surface. Moreover, this anchoring strategy effectively prevents Fe species from clustering or migrating toward the interior of the support during thermal treatment, resulting in atomically dispersed FeN3P-SAC that exhibits a high metallic utilization efficiency and comparable peroxidase-like catalytic activity and kinetics to natural enzymes. As a proof-of-concept demonstration, FeN3P-SAC could effectively block the growth of tumor cells in vitro by combining excellent tumor penetration and the ability to activate chemodynamic and photothermal effects synergistically. This work advances the development of highly active SACs with MPN-based nanotechnology, providing a promising approach for nanocatalytic tumor therapy.
Collapse
Affiliation(s)
- Yajing Zhang
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University Chengdu Sichuan 610065 China
| | - Yunxiang He
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University Chengdu Sichuan 610065 China
| | - Yun Jiao
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University Chengdu Sichuan 610065 China
| | - Guobin Yang
- Weihai Marine Organism & Medical Technology Research Institute, College of Marine Science and Technology, Harbin Institute of Technology Weihai Shandong 264209 China
| | - Yiran Pu
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University Chengdu Sichuan 610065 China
| | - Zhangmin Wan
- Bioproducts Institute, Department of Chemical and Biological Engineering, The University of British Columbia Vancouver BC V6T 1Z4 Canada
| | - Shuyun Li
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University Chengdu Sichuan 610065 China
| | - Yanchao Wu
- Weihai Marine Organism & Medical Technology Research Institute, College of Marine Science and Technology, Harbin Institute of Technology Weihai Shandong 264209 China
| | - Wen Liao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu Sichuan 610041 China
| | - Junling Guo
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University Chengdu Sichuan 610065 China
- Bioproducts Institute, Department of Chemical and Biological Engineering, The University of British Columbia Vancouver BC V6T 1Z4 Canada
- State Key Laboratory of Polymer Materials Engineering, Sichuan University Chengdu Sichuan 610065 China
| |
Collapse
|
3
|
Seo E, Shin EC, Jung MK. SARS-CoV-2 vaccine-elicited immune responses in solid organ transplant recipients. CLINICAL TRANSPLANTATION AND RESEARCH 2024; 38:247-256. [PMID: 39743229 PMCID: PMC11732761 DOI: 10.4285/ctr.24.0062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025]
Abstract
Solid organ transplant recipients (SOTRs) are considered a high-risk group for coronavirus disease 2019 (COVID-19). The adaptive immune responses generated by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination include humoral and cellular immune responses. Most studies on the SARS-CoV-2 vaccine have focused primarily on humoral immunity, but cellular immunity is vital for effectively controlling progression to severe COVID-19. In SOTRs, the vaccine-induced adaptive immune response is significantly attenuated compared to the response in healthy individuals. Nevertheless, vaccinated SOTRs exhibit a reduced rate and severity of SARS-CoV-2 infection. This review aims to provide a concise overview of the current understanding of SARS-CoV-2 vaccine-induced immune responses in SOTRs.
Collapse
Affiliation(s)
- Euri Seo
- The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Korea
| | - Eui-Cheol Shin
- The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Min Kyung Jung
- The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Korea
| |
Collapse
|
4
|
Krishna B, Metaxaki M, Perera M, Wills M, Sithole N. Comparison of different T cell assays for the retrospective determination of SARS-CoV-2 infection. J Gen Virol 2024; 105. [PMID: 39704047 DOI: 10.1099/jgv.0.002055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024] Open
Abstract
It is important to be able to retrospectively determine severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections with high accuracy, both for post-coronavirus disease 2019 (COVID-19) epidemiological studies, and to distinguish between Long COVID and other multi-syndromic diseases that have overlapping symptoms. Although serum antibody levels can be measured to retrospectively diagnose SARS-CoV-2 infections, peptide stimulation of memory T cell responses is a more sensitive approach. This is because robust memory T cells are generated after SARS-CoV-2 infection and persist even after antibodies wane below detectability thresholds. In this study, we compare T cell responses using FluoroSpot-based methods and overnight stimulation of whole blood with SARS-CoV-2 peptides followed by an ELISA. Both approaches have comparable sensitivity and specificity but require different equipment and samples to be used. Furthermore, the elimination of peptides that cross-react with other coronaviruses increases the assay specificity but trades off some sensitivity. Finally, this approach can be used on archival, cryopreserved PBMCs. This work shows comparative advantages for several methods to measure SARS-CoV-2 T cell responses that could be utilized by any laboratory studying the effects of the coronavirus disease 2019 pandemic.
Collapse
Affiliation(s)
- Benjamin Krishna
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
| | - Marina Metaxaki
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
| | - Marianne Perera
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Mark Wills
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
| | - Nyarie Sithole
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
| |
Collapse
|
5
|
Benoit JM, Breznik JA, Huynh A, Cowbrough B, Baker B, Heessels L, Lodhi S, Yan E, Bhakta H, Clare R, Nazy I, Bramson JL, Larché MJ, Bowdish DM. Reassuring humoral and cellular immune responses to SARS-CoV-2 vaccination in participants with systemic sclerosis. Immunol Lett 2024; 270:106929. [PMID: 39305938 DOI: 10.1016/j.imlet.2024.106929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 09/05/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024]
Abstract
Individuals with systemic sclerosis (SSc) are particularly susceptible to SARS-CoV-2 infections, yet it remains to be determined if they generate humoral and cellular responses comparable to controls following SARS-CoV-2 vaccinations. Herein, we collected blood and serum after second, third, and fourth SARS-CoV-2 vaccinations in patients with SSc and controls. Following each dose, participants with SSc mounted comparable serum anti-RBD IgG, anti-RBD IgA, and spike-specific CD4+ and CD8+T cell responses to those found in controls. At 3 months post dose 2, the frequencies of Th1, Th2, Th17, and Treg spike-specific CD4+T cells in participants with SSc did not differ from controls. At 2-6 weeks post dose 3, participants with SSc displayed reduced frequencies, but not numbers, of Th17-polarized spike-specific CD4+T cells. Thus, participants with SSc did not display significantly weaker humoral or cellular responses to SARS-CoV-2 vaccination than controls, enabling reassurance of vaccine immunogenicity in participants with SSc.
Collapse
Affiliation(s)
- Jenna M Benoit
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada; McMaster Immunology Research Centre, Hamilton, Ontario, Canada; Firestone Institute of Respiratory Health, St. Joseph's Healthcare, Hamilton, Ontario, Canada
| | - Jessica A Breznik
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada; McMaster Immunology Research Centre, Hamilton, Ontario, Canada; McMaster Institute for Research on Aging, McMaster University, Hamilton, Ontario, Canada
| | - Angela Huynh
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Braeden Cowbrough
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada; McMaster Immunology Research Centre, Hamilton, Ontario, Canada
| | - Barbara Baker
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Lauren Heessels
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Sumiya Lodhi
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Elizabeth Yan
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Hina Bhakta
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Rumi Clare
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Ishac Nazy
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan L Bramson
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada; McMaster Immunology Research Centre, Hamilton, Ontario, Canada
| | - Maggie J Larché
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| | - Dawn Me Bowdish
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada; McMaster Immunology Research Centre, Hamilton, Ontario, Canada; Firestone Institute of Respiratory Health, St. Joseph's Healthcare, Hamilton, Ontario, Canada.
| |
Collapse
|
6
|
Lee H, Ko GY, Lee J, Bae H, Ryu JH, Jung J, Kang H, Lee R, Lee DG, Oh EJ. Longitudinal Comparison of Three T-Cell Assays and Three Antibody Assays Against SARS-CoV-2 Following Homologous mRNA-1273/mRNA-1273/mRNA-1273 and Heterologous ChAdOx1/ChAdOx1/BNT162b2 Vaccination: A Prospective Cohort in Naïve Healthcare Workers. Vaccines (Basel) 2024; 12:1350. [PMID: 39772013 PMCID: PMC11679843 DOI: 10.3390/vaccines12121350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Cellular and humoral immunity are key to the immune response against SARS-CoV-2, but the comparability and correlation across different assays remain underexplored. This study compares three T-cell and three antibody assays in two vaccine groups. Methods: This prospective longitudinal cohort study involved 46 naïve healthcare workers: a total of 11 in the homologous mRNA-1273 group (three doses) and 35 in the heterologous ChAd group (two ChAd doses followed by a BNT booster). Blood samples were collected at five time points. Cellular immunity was assessed using ELISPOT and two commercial interferon-gamma release assays: (IGRA)-QuantiFERON SARS-CoV-2 (QF) and Covi-FERON ELISA (CoVF). Humoral immunity was evaluated using total and IgG antibody assays and a surrogate virus neutralization test. Results: The mRNA-1273 group exhibited stronger and more consistent responses than the ChAd group. The correlations between ELISPOT and IGRA varied from weak to moderate (ρ = 0.300-0.410), while QF-IGRA and CoVF-IGRA showed stronger correlations (ρ = 0.700-0.737). The ELISPOT assay showed substantial agreement with QF [Ag2]-IGRA (k = 0.697-0.774) and CoVF [O-sp]-IGRA (k = 0.641-0.718), and an 80.4% agreement rate (k = 0.608) was found between the QF [Ag2]- and CoVF [O-sp]-IGRA tests. Three antibody assays demonstrated very strong correlations with each other and substantial to near-perfect agreement with ELISPOT (k = 0.866-0.949), QF [Ag2]-IGRA (k = 0.807-0.831), and CoVF [O-sp]-IGRA (k = 0.753-0.777). Conclusions: SARS-CoV-2-specific cellular and antibody responses vary by platform and vaccine type, highlighting the importance of measuring both T-cell and B-cell responses using multiple assays to comprehensively assess immune status.
Collapse
Affiliation(s)
- Hyeyoung Lee
- Department of Laboratory Medicine, International St. Mary’s Hospital, College of Medicine, Catholic Kwandong University, Incheon 22711, Republic of Korea;
| | - Geon Young Ko
- Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul 06591, Republic of Korea; (G.Y.K.); (J.L.); (H.B.)
| | - Jihyun Lee
- Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul 06591, Republic of Korea; (G.Y.K.); (J.L.); (H.B.)
| | - Hyunjoo Bae
- Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul 06591, Republic of Korea; (G.Y.K.); (J.L.); (H.B.)
| | - Ji Hyeong Ryu
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.H.R.); (J.J.); (H.K.)
| | - Jin Jung
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.H.R.); (J.J.); (H.K.)
| | - Hyunhye Kang
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.H.R.); (J.J.); (H.K.)
- Research and Development Institute for In Vitro Diagnostic Medical Devices, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Raeseok Lee
- Division of Infectious Diseases, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (R.L.); (D.-G.L.)
- Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Dong-Gun Lee
- Division of Infectious Diseases, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (R.L.); (D.-G.L.)
- Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Eun-Jee Oh
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.H.R.); (J.J.); (H.K.)
- Research and Development Institute for In Vitro Diagnostic Medical Devices, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
7
|
Hopkins G, Gomez N, Tucis D, Bartlett L, Steers G, Burns E, Brown M, Harvey-Cowlishaw T, Santos R, Lauder SN, Scurr M, Capitani L, Burnell S, Rees T, Smart K, Somerville M, Gallimore A, Perera M, Potts M, Metaxaki M, Krishna B, Jackson H, Tighe P, Onion D, Godkin A, Wills M, Fairclough L. Lower Humoral and Cellular Immunity Following Asymptomatic SARS-CoV-2 Infection Compared to Symptomatic Infection in Education (The ACE Cohort). J Clin Immunol 2024; 44:147. [PMID: 38856804 PMCID: PMC11164737 DOI: 10.1007/s10875-024-01739-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/20/2024] [Indexed: 06/11/2024]
Abstract
PURPOSE Asymptomatic SARS-CoV-2 infections were widely reported during the COVID-19 pandemic, acting as a hidden source of infection. Many existing studies investigating asymptomatic immunity failed to recruit true asymptomatic individuals. Thus, we conducted a longitudinal cohort study to evaluate humoral- and cell-mediated responses to infection and vaccination in well-defined asymptomatic young adults (the Asymptomatic COVID-19 in Education [ACE] cohort). METHODS Asymptomatic testing services located at three UK universities identified asymptomatic young adults who were subsequently recruited with age- and sex-matched symptomatic and uninfected controls. Blood and saliva samples were collected after SARS-CoV-2 Wuhan infection, and again after vaccination. 51 participant's anti-spike antibody titres, neutralizing antibodies, and spike-specific T-cell responses were measured, against both Wuhan and Omicron B.1.1.529.1. RESULTS Asymptomatic participants exhibited reduced Wuhan-specific neutralization antibodies pre- and post-vaccination, as well as fewer Omicron-specific neutralization antibodies post-vaccination, compared to symptomatic participants. Lower Wuhan and Omicron-specific IgG titres in asymptomatic individuals were also observed pre- and post-vaccination, compared to symptomatic participants. There were no differences in salivary IgA levels. Conventional flow cytometry analysis and multi-dimensional clustering analysis indicated unvaccinated asymptomatic participants had significantly fewer Wuhan-specific IL-2 secreting CD4+ CD45RA+ T cells and activated CD8+ T cells than symptomatic participants, though these differences dissipated after vaccination. CONCLUSIONS Asymptomatic infection results in decreased antibody and T cell responses to further exposure to SARS-CoV-2 variants, compared to symptomatic infection. Post-vaccination, antibody responses are still inferior, but T cell immunity increases to match symptomatic subjects, emphasising the importance of vaccination to help protect asymptomatic individuals against future variants.
Collapse
Affiliation(s)
- Georgina Hopkins
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Nancy Gomez
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Davis Tucis
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Laura Bartlett
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Graham Steers
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Ellie Burns
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Michaela Brown
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | | | - Rute Santos
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | | | - Martin Scurr
- School of Medicine, Cardiff University, Cardiff, UK
- ImmunoServ Ltd, Cardiff, UK
| | | | | | - Tara Rees
- School of Medicine, Cardiff University, Cardiff, UK
| | | | | | | | - Marianne Perera
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Martin Potts
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Marina Metaxaki
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Hannah Jackson
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Paddy Tighe
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - David Onion
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Andrew Godkin
- School of Medicine, Cardiff University, Cardiff, UK
- ImmunoServ Ltd, Cardiff, UK
| | - Mark Wills
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Lucy Fairclough
- School of Life Sciences, University of Nottingham, Nottingham, UK.
| |
Collapse
|
8
|
Yang M, Meng Y, Hao W, Zhang J, Liu J, Wu L, Lin B, Liu Y, Zhang Y, Yu X, Wang X, Gong Y, Ge L, Fan Y, Xie C, Xu Y, Chang Q, Zhang Y, Qin X. A prognostic model for SARS-CoV-2 breakthrough infection: Analyzing a prospective cellular immunity cohort. Int Immunopharmacol 2024; 131:111829. [PMID: 38489974 DOI: 10.1016/j.intimp.2024.111829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/03/2024] [Accepted: 03/06/2024] [Indexed: 03/17/2024]
Abstract
BACKGROUND Following the COVID-19 pandemic, studies have identified several prevalent characteristics, especially related to lymphocyte subsets. However, limited research is available on the focus of this study, namely, the specific memory cell subsets among individuals who received COVID-19 vaccine boosters and subsequently experienced a SARS-CoV-2 breakthrough infection. METHODS Flow cytometry (FCM) was employed to investigate the early and longitudinal pattern changes of cellular immunity in patients with SARS-CoV-2 breakthrough infections following COVID-19 vaccine boosters. XGBoost (a machine learning algorithm) was employed to analyze cellular immunity prior to SARS-CoV-2 breakthrough, aiming to establish a prognostic model for SARS-CoV-2 breakthrough infections. RESULTS Following SARS-CoV-2 breakthrough infection, naïve T cells and TEMRA subsets increased while the percentage of TCM and TEM cells decreased. Naïve and non-switched memory B cells increased while switched and double-negative memory B cells decreased. The XGBoost model achieved an area under the curve (AUC) of 0.78, with an accuracy rate of 81.8 %, a sensitivity of 75 %, and specificity of 85.7 %. TNF-α, CD27-CD19+cells, and TEMRA subsets were identified as high predictors. An increase in TNF-α, cTfh, double-negative memory B cells, IL-6, IL-10, and IFN-γ prior to SARS-CoV-2 infection was associated with enduring clinical symptoms; conversely, an increase in CD3+ T cells, CD4+ T cells, and IL-2 was associated with clinical with non-enduring clinical symptoms. CONCLUSION SARS-CoV-2 breakthrough infection leads to disturbances in cellular immunity. Assessing cellular immunity prior to breakthrough infection serves as a valuable prognostic tool for SARS-CoV-2 infection, which facilitates clinical decision-making.
Collapse
Affiliation(s)
- Mei Yang
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yuan Meng
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Wudi Hao
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Jin Zhang
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Jianhua Liu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Lina Wu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Baoxu Lin
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yong Liu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yue Zhang
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xiaojun Yu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xiaoqian Wang
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yu Gong
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Lili Ge
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yan Fan
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Conghong Xie
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yiyun Xu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Qing Chang
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yixiao Zhang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| | - Xiaosong Qin
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
9
|
Cankat S, Demael MU, Swadling L. In search of a pan-coronavirus vaccine: next-generation vaccine design and immune mechanisms. Cell Mol Immunol 2024; 21:103-118. [PMID: 38148330 PMCID: PMC10805787 DOI: 10.1038/s41423-023-01116-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 11/21/2023] [Indexed: 12/28/2023] Open
Abstract
Members of the coronaviridae family are endemic to human populations and have caused several epidemics and pandemics in recent history. In this review, we will discuss the feasibility of and progress toward the ultimate goal of creating a pan-coronavirus vaccine that can protect against infection and disease by all members of the coronavirus family. We will detail the unmet clinical need associated with the continued transmission of SARS-CoV-2, MERS-CoV and the four seasonal coronaviruses (HCoV-OC43, NL63, HKU1 and 229E) in humans and the potential for future zoonotic coronaviruses. We will highlight how first-generation SARS-CoV-2 vaccines and natural history studies have greatly increased our understanding of effective antiviral immunity to coronaviruses and have informed next-generation vaccine design. We will then consider the ideal properties of a pan-coronavirus vaccine and propose a blueprint for the type of immunity that may offer cross-protection. Finally, we will describe a subset of the diverse technologies and novel approaches being pursued with the goal of developing broadly or universally protective vaccines for coronaviruses.
Collapse
Affiliation(s)
- S Cankat
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London, NW3 2PP, UK
| | - M U Demael
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London, NW3 2PP, UK
| | - L Swadling
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London, NW3 2PP, UK.
| |
Collapse
|
10
|
Thomas Κ, Grigoropoulos I, Alexopoulou P, Karofylakis E, Galani I, Papadopoulou KK, Tsiavou A, Ntourou A, Mavrou E, Qevani I, Katsimbri P, Koutsianas C, Mavrea E, Vassilopoulos D, Pournaras S, Tsiodras S, Boumpas D, Antoniadou A. Sustained cell-mediated but not humoral responses in rituximab-treated rheumatic patients after vaccination against SARS-CoV-2. Rheumatology (Oxford) 2024; 63:534-541. [PMID: 37228039 PMCID: PMC10836975 DOI: 10.1093/rheumatology/kead236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 04/18/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023] Open
Abstract
OBJECTIVES B-cell depleting monoclonal antibodies are associated with increased COVID-19 severity and impaired immune response to vaccination. We aimed to assess the humoral and cell mediated (CMI) immune response after SARS-CoV-2 vaccination in rituximab (RTX)-treated rheumatic patients. METHODS Serum and whole blood samples were collected from RTX-treated rheumatic patients 3-6 months after last vaccination against SARS-CoV-2. Serum was tested by ELISA for quantitative detection of anti-spike SARS-CoV-2 IgG. Cell-mediated variant-specific SARS-CoV-2 immunity (CMI) was assessed by interferon-γ release assay Covi-FERON FIA. Patients were interviewed for breakthrough COVID-19 infection (BTI) 3 months post sampling. RESULTS Sixty patients were studied after a median (IQR) of 179 (117-221.5) days from last vaccine to sampling. Forty (66.7%) patients had positive Covi-FERON and 23 (38.3%) had detectable anti-spike IgG. Covi-FERON positive patients had lower median RTX cumulative dose [6 (4-10.75) vs 11 (6.75-14.75) grams, (P = 0.019)]. Patients with positive anti-spike IgG had received fewer RTX cycles [2 (2-4) vs 6 (4-8), P = 0.002] and cumulative dose [4 (3-7) vs 10 (6.25-13) grams, P = 0.002] and had shorter time from last vaccination to sampling [140 (76-199) vs 192 (128-230) days, P = 0.047]. Thirty-seven percent were positive only for Covi-FERON and 7% only for anti-spike IgG. Twenty (33.3%) BTI occurred post sampling, exclusively during Omicron variant predominance. The proportion of patients with CMI response against Delta variant was lower in patients who experienced BTI (25% vs 55%, P = 0.03). CONCLUSIONS Four out of ten RTX-treated vaccinated patients show lasting cell-mediated immune response despite undetectable anti-spike antibodies. Cumulative RTX dose affects both humoral and cell-mediated responses to SARS-CoV-2 vaccines. Cell-mediated immune responses call for attention as a vaccine efficacy marker against SARS-CoV-2.
Collapse
Affiliation(s)
- Κonstantinos Thomas
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Attikon University General Hospital, Chaidari, Greece
| | - Ioannis Grigoropoulos
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Attikon University General Hospital, Chaidari, Greece
| | - Panagiota Alexopoulou
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Attikon University General Hospital, Chaidari, Greece
| | - Emmanouil Karofylakis
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Attikon University General Hospital, Chaidari, Greece
| | - Irene Galani
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Attikon University General Hospital, Chaidari, Greece
| | - Kyriaki Korina Papadopoulou
- Clinical Microbiology Laboratory, National and Kapodistrian University of Athens School of Medicine, Attikon University General Hospital, Chaidari, Greece
| | - Anastasia Tsiavou
- Clinical Microbiology Laboratory, National and Kapodistrian University of Athens School of Medicine, Attikon University General Hospital, Chaidari, Greece
| | - Aliki Ntourou
- Clinical Immunology-Rheumatology Unit, 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Attikon University General Hospital, Chaidari, Greece
| | - Eleftheria Mavrou
- Clinical Immunology-Rheumatology Unit, 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Attikon University General Hospital, Chaidari, Greece
| | - Irina Qevani
- Clinical Immunology-Rheumatology Unit, 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Attikon University General Hospital, Chaidari, Greece
| | - Pelagia Katsimbri
- Clinical Immunology-Rheumatology Unit, 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Attikon University General Hospital, Chaidari, Greece
| | - Christos Koutsianas
- Clinical Immunology-Rheumatology Unit, 2nd Department of Medicine and Laboratory, National and Kapodistrian University of Athens School of Medicine, Hippokration General Hospital, Athens, Greece
| | - Evgenia Mavrea
- Clinical Immunology-Rheumatology Unit, 2nd Department of Medicine and Laboratory, National and Kapodistrian University of Athens School of Medicine, Hippokration General Hospital, Athens, Greece
| | - Dimitrios Vassilopoulos
- Clinical Immunology-Rheumatology Unit, 2nd Department of Medicine and Laboratory, National and Kapodistrian University of Athens School of Medicine, Hippokration General Hospital, Athens, Greece
| | - Spyros Pournaras
- Clinical Microbiology Laboratory, National and Kapodistrian University of Athens School of Medicine, Attikon University General Hospital, Chaidari, Greece
| | - Sotirios Tsiodras
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Attikon University General Hospital, Chaidari, Greece
| | - Dimitrios Boumpas
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Attikon University General Hospital, Chaidari, Greece
- Clinical Immunology-Rheumatology Unit, 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Attikon University General Hospital, Chaidari, Greece
| | - Anastasia Antoniadou
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Attikon University General Hospital, Chaidari, Greece
| |
Collapse
|
11
|
Saito T, Couzinet A, Murakami T, Shimomura M, Suzuki T, Katayama Y, Nakatsura T. Rapid and high throughput assessment of cellular immunity against SARS-CoV-2 based on the ex vivo activation of genes in leukocyte assay with whole blood. Biochem Biophys Res Commun 2024; 694:149398. [PMID: 38134475 DOI: 10.1016/j.bbrc.2023.149398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 12/05/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023]
Abstract
During the novel coronavirus outbreak and vaccine development, antibody production garnered major focus as the primary immunogenic response. However, cellular immunity's recent demonstration of comparable or greater significance in controlling infection demands the re-evaluation of the importance of T-cell immunity in SARS-CoV-2 infection. Here, we developed a novel assay, the ex vivo activation of genes in leukocytes (EAGL), which employs short-term whole blood stimulation with the LeukoComplete™ system, to measure ex vivo SARS-CoV-2-specific T cell responses (cellular immunity). This assay measures upregulated mRNA expression related to leukocyte activation 4 h after antigen stimulation. LeukoComplete™ system uses whole blood samples, eliminating the need for pretreatment before analysis. Furthermore, this system's high reproducibility is ensured through a series of operations from mRNA extraction to cDNA synthesis on a 96-well plate. In the performance evaluation using fresh blood from previously SARS-CoV-2-infected and COVID-19-vaccinated individuals, the EAGL assay had a comparable sensitivity and specificity to the ELISpot assay (EAGL: 1.000/1.000; ELISpot: 0.900/0.973). As a simple, high-throughput assay, the EAGL assay is also a quantitative test that is useful in studies with large sample numbers, such as monitoring new vaccine efficacies against novel coronaviruses or epidemiologic studies that require cellular immune testing during viral infection.
Collapse
Affiliation(s)
- Taro Saito
- Minaris Medical Co., Ltd, Nagaizumi, Shizuoka, 411-0932, Japan
| | - Arnaud Couzinet
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, 277-8577, Japan
| | | | - Manami Shimomura
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, 277-8577, Japan
| | - Toshihiro Suzuki
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, 277-8577, Japan
| | - Yuki Katayama
- Minaris Medical Co., Ltd, Nagaizumi, Shizuoka, 411-0932, Japan; Resonac Corporation, Minato, Tokyo, 105-7325, Japan.
| | - Tetsuya Nakatsura
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, 277-8577, Japan
| |
Collapse
|
12
|
Binayke A, Zaheer A, Vishwakarma S, Singh S, Sharma P, Chandwaskar R, Gosain M, Raghavan S, Murugesan DR, Kshetrapal P, Thiruvengadam R, Bhatnagar S, Pandey AK, Garg PK, Awasthi A. A quest for universal anti-SARS-CoV-2 T cell assay: systematic review, meta-analysis, and experimental validation. NPJ Vaccines 2024; 9:3. [PMID: 38167915 PMCID: PMC10762233 DOI: 10.1038/s41541-023-00794-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
Measuring SARS-CoV-2-specific T cell responses is crucial to understanding an individual's immunity to COVID-19. However, high inter- and intra-assay variability make it difficult to define T cells as a correlate of protection against COVID-19. To address this, we performed systematic review and meta-analysis of 495 datasets from 94 original articles evaluating SARS-CoV-2-specific T cell responses using three assays - Activation Induced Marker (AIM), Intracellular Cytokine Staining (ICS), and Enzyme-Linked Immunospot (ELISPOT), and defined each assay's quantitative range. We validated these ranges using samples from 193 SARS-CoV-2-exposed individuals. Although IFNγ ELISPOT was the preferred assay, our experimental validation suggested that it under-represented the SARS-CoV-2-specific T cell repertoire. Our data indicate that a combination of AIM and ICS or FluoroSpot assay would better represent the frequency, polyfunctionality, and compartmentalization of the antigen-specific T cell responses. Taken together, our results contribute to defining the ranges of antigen-specific T cell assays and propose a choice of assay that can be employed to better understand the cellular immune response against viral diseases.
Collapse
Affiliation(s)
- Akshay Binayke
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, Faridabad, India
- Jawaharlal Nehru University, New Delhi, India
| | - Aymaan Zaheer
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Siddhesh Vishwakarma
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Savita Singh
- Translational Health Science and Technology Institute, Faridabad, India
| | - Priyanka Sharma
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Rucha Chandwaskar
- Department of Microbiology, AMITY University Rajasthan, Jaipur, India
| | - Mudita Gosain
- Translational Health Science and Technology Institute, Faridabad, India
| | | | | | | | - Ramachandran Thiruvengadam
- Translational Health Science and Technology Institute, Faridabad, India
- Pondicherry Institute of Medical Sciences, Puducherry, India
| | | | | | - Pramod Kumar Garg
- Translational Health Science and Technology Institute, Faridabad, India
- All India Institute of Medical Sciences, New Delhi, India
| | - Amit Awasthi
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India.
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, Faridabad, India.
| |
Collapse
|
13
|
Murray SM, Pose E, Wittner M, Londoño MC, Schaub G, Cook J, Dimitriadis S, Meacham G, Irwin S, Lim Z, Duengelhoef P, Sterneck M, Lohse AW, Perez V, Trivedi P, Bhandal K, Mullish B, Manousou P, Provine NM, Avitabile E, Carroll M, Tipton T, Healy S, Burra P, Klenerman P, Dunachie S, Kronsteiner B, Maciola AK, Pasqual G, Hernandez-Gea V, Garcia-Pagan JC, Lampertico P, Iavarone M, Gines P, Lütgehetmann M, Schulze Zur Wiesch J, Russo FP, Barnes E, Marjot T, Middleton G, Gaskell C, Rea D, Pirrie S, Bowden SJ, Pope A, Hughes A, Harrison M, Kirkham A, Middleton L, Lowe F, Magwaro S, Kearns P, Lim SH, Willicombe M, Prendecki M, Clarke C, Mortimer P, McIntyre S, Thomas D, Richter A, Al-Taei S, Goodyear CS, Siebert S, Basu N, Gilmour A, McInnes IB, Tong A, Woolcock K, Basheer F, Crawley C, Malladi R, King A, Lockey S, Uttenthal B, Snowden JA, Selby R, Orchard K, de Silva TI, Meardon N, Hansford S, Sandhar G, Kelleher P, Kesavan M, Moore C, Manousou P, Hahn G, Mullish B, Atta M, Gleeson S, Lightstone L, Martin P, McAdoo S, Thomson T, Koh MB, Avenoso D, Sanderson R, Taylor C, Bhandal K, Hall D, Filer A, et alMurray SM, Pose E, Wittner M, Londoño MC, Schaub G, Cook J, Dimitriadis S, Meacham G, Irwin S, Lim Z, Duengelhoef P, Sterneck M, Lohse AW, Perez V, Trivedi P, Bhandal K, Mullish B, Manousou P, Provine NM, Avitabile E, Carroll M, Tipton T, Healy S, Burra P, Klenerman P, Dunachie S, Kronsteiner B, Maciola AK, Pasqual G, Hernandez-Gea V, Garcia-Pagan JC, Lampertico P, Iavarone M, Gines P, Lütgehetmann M, Schulze Zur Wiesch J, Russo FP, Barnes E, Marjot T, Middleton G, Gaskell C, Rea D, Pirrie S, Bowden SJ, Pope A, Hughes A, Harrison M, Kirkham A, Middleton L, Lowe F, Magwaro S, Kearns P, Lim SH, Willicombe M, Prendecki M, Clarke C, Mortimer P, McIntyre S, Thomas D, Richter A, Al-Taei S, Goodyear CS, Siebert S, Basu N, Gilmour A, McInnes IB, Tong A, Woolcock K, Basheer F, Crawley C, Malladi R, King A, Lockey S, Uttenthal B, Snowden JA, Selby R, Orchard K, de Silva TI, Meardon N, Hansford S, Sandhar G, Kelleher P, Kesavan M, Moore C, Manousou P, Hahn G, Mullish B, Atta M, Gleeson S, Lightstone L, Martin P, McAdoo S, Thomson T, Koh MB, Avenoso D, Sanderson R, Taylor C, Bhandal K, Hall D, Filer A, Trivedi P, Cook G, Hurst E, Publicover A, Scouse K, Klenerman P, Dunachie SJ, Barnes E, Murray SM, Lim Z, Satsangi J, Irwin S, Meacham G, Marjot T, Dimitriadis S, Chalk J, Hanke D, Hanke J, Healy S, Laidlaw S, Longet S, Provine N, Thomas S, Walker V, Win Z, Beesley R, Churchill V, Loughton H, Insch E, MacDonald E, Trown D, Faria P, Chackathayil J, Hutchison C, Richardson D, Arnott M, Bennett L, Brock J, Keillor V, Melville A, Melville L, Miller S, Najm A, Paterson C, Rodgers L, Rutherford M, Rundell S, Smith E, Stewart L, Sunzini F, Carroll M. Immune responses and clinical outcomes after COVID-19 vaccination in patients with liver disease and liver transplant recipients. J Hepatol 2024; 80:109-123. [PMID: 37863203 PMCID: PMC10914634 DOI: 10.1016/j.jhep.2023.10.009] [Show More Authors] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/20/2023] [Accepted: 10/03/2023] [Indexed: 10/22/2023]
Abstract
BACKGROUND & AIMS Comparative assessments of immunogenicity following different COVID-19 vaccines in patients with distinct liver diseases are lacking. SARS-CoV-2-specific T-cell and antibody responses were evaluated longitudinally after one to three vaccine doses, with long-term follow-up for COVID-19-related clinical outcomes. METHODS A total of 849 participants (355 with cirrhosis, 74 with autoimmune hepatitis [AIH], 36 with vascular liver disease [VLD], 257 liver transplant recipients [LTRs] and 127 healthy controls [HCs]) were recruited from four countries. Standardised immune assays were performed pre and post three vaccine doses (V1-3). RESULTS In the total cohort, there were incremental increases in antibody titres after each vaccine dose (p <0.0001). Factors associated with reduced antibody responses were age and LT, whereas heterologous vaccination, prior COVID-19 and mRNA platforms were associated with greater responses. Although antibody titres decreased between post-V2 and pre-V3 (p = 0.012), patients with AIH, VLD, and cirrhosis had equivalent antibody responses to HCs post-V3. LTRs had lower and more heterogenous antibody titres than other groups, including post-V3 where 9% had no detectable antibodies; this was heavily influenced by intensity of immunosuppression. Vaccination increased T-cell IFNγ responses in all groups except LTRs. Patients with liver disease had lower functional antibody responses against nine Omicron subvariants and reduced T-cell responses to Omicron BA.1-specific peptides compared to wild-type. 122 cases of breakthrough COVID-19 were reported of which 5/122 (4%) were severe. Of the severe cases, 4/5 (80%) occurred in LTRs and 2/5 (40%) had no serological response post-V2. CONCLUSION After three COVID-19 vaccines, patients with liver disease generally develop robust antibody and T-cell responses to vaccination and have mild COVID-19. However, LTRs have sustained no/low antibody titres and appear most vulnerable to severe disease. IMPACT AND IMPLICATIONS Standardised assessments of the immune response to different COVID-19 vaccines in patients with liver disease are lacking. We performed antibody and T-cell assays at multiple timepoints following up to three vaccine doses in a large cohort of patients with a range of liver conditions. Overall, the three most widely available vaccine platforms were immunogenic and appeared to protect against severe breakthrough COVID-19. This will provide reassurance to patients with chronic liver disease who were deemed at high risk of severe COVID-19 during the pre-vaccination era, however, liver transplant recipients had the lowest antibody titres and remained vulnerable to severe breakthrough infection. We also characterise the immune response to multiple SARS-CoV-2 variants and describe the interaction between disease type, severity, and vaccine platform. These insights may prove useful in the event of future viral infections which also require rapid vaccine development and delivery to patients with liver disease.
Collapse
Affiliation(s)
- Sam M Murray
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Elisa Pose
- Liver Unit, Hospital Clínic, Institut de Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain; CIBERehd (Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas), Spain
| | - Melanie Wittner
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Germany; Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maria-Carlota Londoño
- Liver Unit, Hospital Clínic, Institut de Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain; CIBERehd (Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas), Spain
| | - Golda Schaub
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Germany; Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jonathan Cook
- Centre for Statistics in Medicine, University of Oxford, Oxford, UK
| | - Stavros Dimitriadis
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Georgina Meacham
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sophie Irwin
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Zixiang Lim
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Paul Duengelhoef
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martina Sterneck
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ansgar W Lohse
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Germany; Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Valeria Perez
- Liver Unit, Hospital Clínic, Institut de Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain; CIBERehd (Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas), Spain; Health Care Provider of the European Reference Network on Rare Liver Disorders (ERN-Liver)
| | - Palak Trivedi
- National Institute for Health Research Birmingham Biomedical Research Centre, Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, UK; Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Birmingham, UK
| | - Khush Bhandal
- Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Birmingham, UK
| | - Ben Mullish
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK; Department of Hepatology, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Pinelopi Manousou
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK; Department of Hepatology, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Nicholas M Provine
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Emma Avitabile
- Liver Unit, Hospital Clínic, Institut de Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain; CIBERehd (Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas), Spain
| | - Miles Carroll
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Tom Tipton
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Saoirse Healy
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Patrizia Burra
- University of Padova, Department of Surgery, Oncology and Gastroenterology DISCOG, Italy
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK; The Oxford NIHR Biomedical Research Centre, Oxford University Hospital NHS Trust, Oxford, UK
| | - Susanna Dunachie
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; The Oxford NIHR Biomedical Research Centre, Oxford University Hospital NHS Trust, Oxford, UK; Mahidol Oxford Tropical Medicine Research Unit, University of Mahidol, Bangkok, Thailand
| | - Barbara Kronsteiner
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; The Oxford NIHR Biomedical Research Centre, Oxford University Hospital NHS Trust, Oxford, UK; Mahidol Oxford Tropical Medicine Research Unit, University of Mahidol, Bangkok, Thailand
| | - Agnieszka Katarzyna Maciola
- Laboratory of Synthetic Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Giulia Pasqual
- Laboratory of Synthetic Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Virginia Hernandez-Gea
- Liver Unit, Hospital Clínic, Institut de Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain; CIBERehd (Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas), Spain; Health Care Provider of the European Reference Network on Rare Liver Disorders (ERN-Liver)
| | - Juan Carlos Garcia-Pagan
- Liver Unit, Hospital Clínic, Institut de Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain; CIBERehd (Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas), Spain; Health Care Provider of the European Reference Network on Rare Liver Disorders (ERN-Liver)
| | - Pietro Lampertico
- Division of Gastroenterology and Hepatology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; CRC "A. M. and A. Migliavacca" Center for Liver Disease, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Massimo Iavarone
- Division of Gastroenterology and Hepatology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Pere Gines
- Liver Unit, Hospital Clínic, Institut de Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain; CIBERehd (Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas), Spain
| | - Marc Lütgehetmann
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julian Schulze Zur Wiesch
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Germany; Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Francesco Paolo Russo
- University of Padova, Department of Surgery, Oncology and Gastroenterology DISCOG, Italy
| | - Eleanor Barnes
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; The Oxford NIHR Biomedical Research Centre, Oxford University Hospital NHS Trust, Oxford, UK.
| | - Thomas Marjot
- Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, Churchill Hospital, University of Oxford, Oxford, UK; Oxford Liver Unit, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK.
| | - Gary Middleton
- Cancer Centre, University Hospitals Birmingham, NHS Foundation Trust, Birmingham B15 2WB, UK
| | - Charlotte Gaskell
- Cancer Centre, University Hospitals Birmingham, NHS Foundation Trust, Birmingham B15 2WB, UK
| | - Daniel Rea
- Cancer Centre, University Hospitals Birmingham, NHS Foundation Trust, Birmingham B15 2WB, UK
| | - Sarah Pirrie
- Cancer Centre, University Hospitals Birmingham, NHS Foundation Trust, Birmingham B15 2WB, UK
| | - Sarah J Bowden
- Cancer Centre, University Hospitals Birmingham, NHS Foundation Trust, Birmingham B15 2WB, UK
| | - Ann Pope
- Cancer Centre, University Hospitals Birmingham, NHS Foundation Trust, Birmingham B15 2WB, UK
| | - Ana Hughes
- Cancer Centre, University Hospitals Birmingham, NHS Foundation Trust, Birmingham B15 2WB, UK
| | - Molly Harrison
- Cancer Centre, University Hospitals Birmingham, NHS Foundation Trust, Birmingham B15 2WB, UK
| | - Amanda Kirkham
- Cancer Centre, University Hospitals Birmingham, NHS Foundation Trust, Birmingham B15 2WB, UK
| | - Lucinda Middleton
- Cancer Centre, University Hospitals Birmingham, NHS Foundation Trust, Birmingham B15 2WB, UK
| | - Faye Lowe
- Cancer Centre, University Hospitals Birmingham, NHS Foundation Trust, Birmingham B15 2WB, UK
| | - Sophia Magwaro
- Cancer Centre, University Hospitals Birmingham, NHS Foundation Trust, Birmingham B15 2WB, UK
| | - Pamela Kearns
- National Institute for Health Research Birmingham Biomedical Research Centre, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2TT UK; Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Edgbaston, Birmingham. B15 2TT, UK
| | - Sean H Lim
- Centre for Cancer Immunology, University of Southampton, Southampton, SO16 6YD UK
| | - Michelle Willicombe
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN UK
| | - Maria Prendecki
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN UK
| | - Candice Clarke
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN UK
| | - Paige Mortimer
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN UK
| | - Stacey McIntyre
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN UK
| | - David Thomas
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN UK
| | - Alex Richter
- Clinical Immunology Service, University of Birmingham, Edgbaston, Birmingham. B15 2TT, UK
| | - Sally Al-Taei
- Clinical Immunology Service, University of Birmingham, Edgbaston, Birmingham. B15 2TT, UK
| | - Carl S Goodyear
- College of Medical, Veterinary & Life Sciences; University of Glasgow, Glasgow; G12 8QQ, UK
| | - Stefan Siebert
- College of Medical, Veterinary & Life Sciences; University of Glasgow, Glasgow; G12 8QQ, UK
| | - Neil Basu
- College of Medical, Veterinary & Life Sciences; University of Glasgow, Glasgow; G12 8QQ, UK
| | - Ashley Gilmour
- College of Medical, Veterinary & Life Sciences; University of Glasgow, Glasgow; G12 8QQ, UK
| | - Iain B McInnes
- College of Medical, Veterinary & Life Sciences; University of Glasgow, Glasgow; G12 8QQ, UK
| | - Andrew Tong
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 OQQ, UK
| | - Kieran Woolcock
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 OQQ, UK
| | - Faisal Basheer
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 OQQ, UK
| | - Charles Crawley
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 OQQ, UK
| | - Ram Malladi
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 OQQ, UK
| | - Andrew King
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 OQQ, UK
| | - Sophie Lockey
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 OQQ, UK
| | - Ben Uttenthal
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 OQQ, UK
| | - John A Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield. S10 2JF, UK
| | - Rachael Selby
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield. S10 2JF, UK
| | - Kim Orchard
- Department of Haematology, University Hospital Southampton NHS Foundation Trust, Southampton, S016 6YD UK
| | - Thushan I de Silva
- Department of Infection, Immunity and Cardiovascular Disease, The Medical School, The University of Sheffield, Sheffield. S10 2RX, UK
| | - Naomi Meardon
- Department of Infection, Immunity and Cardiovascular Disease, The Medical School, The University of Sheffield, Sheffield. S10 2RX, UK
| | - Sam Hansford
- Department of Infection, Immunity and Cardiovascular Disease, The Medical School, The University of Sheffield, Sheffield. S10 2RX, UK
| | - Gurjinder Sandhar
- Department of Infection, Immunity and Cardiovascular Disease, The Medical School, The University of Sheffield, Sheffield. S10 2RX, UK
| | - Peter Kelleher
- Department of Infectious Diseases, Imperial College London, School of Medicine Chelsea and Westminster Hospital, London SW10 9NH UK
| | - Murali Kesavan
- Department of Oncology, Cancer and Haematology Centre, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LE, UK
| | - Celia Moore
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, W2 1NY, UK
| | - Pinelopi Manousou
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, W2 1NY, UK
| | - Gareth Hahn
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, W2 1NY, UK
| | - Benjamin Mullish
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, W2 1NY, UK
| | - Maria Atta
- Haematology Department, Hammermith Hospital, London, W12 0HS UK
| | - Sarah Gleeson
- Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, W12 OHS, UK
| | - Liz Lightstone
- Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, W12 OHS, UK
| | - Paul Martin
- Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, W12 OHS, UK
| | - Stephen McAdoo
- Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, W12 OHS, UK
| | - Tina Thomson
- Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, W12 OHS, UK
| | - Mickey Bc Koh
- Infection and Immunity Clinical Academic Group, St George's, University of London; Department of Haematology, St George's University Hospital NHS Foundation Trust, London SW17 0QT
| | - Daniele Avenoso
- King's College Hospital NHS Foundation Trust, London, SE5 9RS, UK
| | - Robin Sanderson
- King's College Hospital NHS Foundation Trust, London, SE5 9RS, UK
| | - Claire Taylor
- Leeds Institute of Medical Research, University of Leeds, Leeds, LS2 9NL
| | - Khushpreet Bhandal
- Liver Research Delivery Team, University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2GW, UK
| | - Diana Hall
- Liver Research Delivery Team, University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2GW, UK
| | - Andrew Filer
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre and NIHR Clinical Research Facility, Institute of Inflammation and Ageing, University of Birmingham and University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2TT, UK
| | - Palak Trivedi
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre and NIHR Clinical Research Facility, Institute of Inflammation and Ageing, University of Birmingham and University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2TT, UK
| | - Gordon Cook
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre and NIHR Clinical Research Facility, Institute of Inflammation and Ageing, University of Birmingham and University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2TT, UK
| | - Erin Hurst
- Northern Centre for Cancer Care, Freeman Hospital, Newcastle upon Tyne, NE7 7DN, UK
| | - Amy Publicover
- Northern Centre for Cancer Care, Freeman Hospital, Newcastle upon Tyne, NE7 7DN, UK
| | - Katy Scouse
- Northern Centre for Cancer Care, Freeman Hospital, Newcastle upon Tyne, NE7 7DN, UK
| | - Paul Klenerman
- Nuffield Department of Medicine, University of Oxford, Oxford, OX1 2JD, UK; National Institute for Health Research, Oxford Biomedical Research Centre, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Susanna J Dunachie
- Nuffield Department of Medicine, University of Oxford, Oxford, OX1 2JD, UK; National Institute for Health Research, Oxford Biomedical Research Centre, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Eleanor Barnes
- Nuffield Department of Medicine, University of Oxford, Oxford, OX1 2JD, UK; National Institute for Health Research, Oxford Biomedical Research Centre, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Sam M Murray
- Nuffield Department of Medicine, University of Oxford, Oxford, OX1 2JD, UK
| | - Zixiang Lim
- Nuffield Department of Medicine, University of Oxford, Oxford, OX1 2JD, UK
| | - Jack Satsangi
- Nuffield Department of Medicine, University of Oxford, Oxford, OX1 2JD, UK
| | - Sophie Irwin
- Nuffield Department of Medicine, University of Oxford, Oxford, OX1 2JD, UK
| | - Georgina Meacham
- Nuffield Department of Medicine, University of Oxford, Oxford, OX1 2JD, UK
| | - Thomas Marjot
- Nuffield Department of Medicine, University of Oxford, Oxford, OX1 2JD, UK
| | | | - Jem Chalk
- Nuffield Department of Medicine, University of Oxford, Oxford, OX1 2JD, UK
| | - Daniel Hanke
- Nuffield Department of Medicine, University of Oxford, Oxford, OX1 2JD, UK
| | - Josef Hanke
- Nuffield Department of Medicine, University of Oxford, Oxford, OX1 2JD, UK
| | - Saoirse Healy
- Nuffield Department of Medicine, University of Oxford, Oxford, OX1 2JD, UK
| | - Stephen Laidlaw
- Nuffield Department of Medicine, University of Oxford, Oxford, OX1 2JD, UK
| | - Stephanie Longet
- Nuffield Department of Medicine, University of Oxford, Oxford, OX1 2JD, UK
| | - Nicholas Provine
- Nuffield Department of Medicine, University of Oxford, Oxford, OX1 2JD, UK
| | - Sarah Thomas
- Nuffield Department of Medicine, University of Oxford, Oxford, OX1 2JD, UK
| | - Victoria Walker
- Nuffield Department of Medicine, University of Oxford, Oxford, OX1 2JD, UK
| | - Zay Win
- Nuffield Department of Medicine, University of Oxford, Oxford, OX1 2JD, UK
| | - Richard Beesley
- Patient and Public Representatives on the Trial Management Group
| | - Vicky Churchill
- Patient and Public Representatives on the Trial Management Group
| | - Holly Loughton
- Patient and Public Representatives on the Trial Management Group
| | - Elspeth Insch
- Patient and Public Representatives on the Trial Management Group
| | - Eilean MacDonald
- Patient and Public Representatives on the Trial Management Group
| | - Doreen Trown
- Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield S10 2JF, UK
| | - Patricia Faria
- St George's hospital and Medical School, St George's University Hospitals NHS Foundation Trust, London, SW17 0QT, UK
| | - Julie Chackathayil
- St George's hospital and Medical School, St George's University Hospitals NHS Foundation Trust, London, SW17 0QT, UK
| | - Clare Hutchison
- University Hospital Southampton NHS Foundation Trust, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Deborah Richardson
- University Hospital Southampton NHS Foundation Trust, Southampton General Hospital, Southampton, SO16 6YD, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Miles Carroll
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
14
|
Benoit JM, Breznik JA, Ang JC, Bhakta H, Huynh A, Cowbrough B, Baker B, Heessels L, Lodhi S, Yan E, Ewusie J, Nazy I, Bramson J, Miller MS, Bernatsky S, Larché MJ, Bowdish DME. Immunomodulatory drugs have divergent effects on humoral and cellular immune responses to SARS-CoV-2 vaccination in people living with rheumatoid arthritis. Sci Rep 2023; 13:22846. [PMID: 38129594 PMCID: PMC10739702 DOI: 10.1038/s41598-023-50263-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023] Open
Abstract
Understanding the efficacy of SARS-CoV-2 vaccination in people on immunosuppressive drugs, including those with rheumatoid arthritis (RA), is critical for their protection. Vaccine induced protection requires antibodies, CD4+ T cells, and CD8+ T cells, but it is unclear if these are equally affected by immunomodulatory drugs. Here, we determined how humoral and cellular SARS-CoV-2 vaccination responses differed between people with RA and controls, and which drug classes impacted these responses. Blood was collected from participants with RA on immunomodulatory drugs and controls after their second, third, and fourth SARS-CoV-2 vaccinations. Receptor binding domain (RBD)-specific antibodies were quantified by ELISA. Spike-specific memory T cells were quantitated using flow cytometry. Linear mixed models assessed the impact of age, sex, and immunomodulatory drug classes on SARS-CoV-2 vaccination responses. Compared to non-RA controls (n = 35), participants with RA on immunomodulatory drugs (n = 62) had lower anti-RBD IgG and spike-specific CD4+ T cell levels, but no deficits in spike-specific CD8+ T cells, following SARS-CoV-2 vaccination. Use of costimulation inhibitors was associated with lower humoral responses. JAK inhibitors were associated with fewer spike-specific CD4+ T cells. Participants with RA on immunomodulatory drugs mounted weaker responses to SARS-CoV-2 vaccination, with different drug classes impacting the cellular and humoral compartments.
Collapse
Affiliation(s)
- Jenna M Benoit
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, Hamilton, ON, Canada
- Firestone Institute of Respiratory Health, St. Joseph's Healthcare, Hamilton, ON, Canada
| | - Jessica A Breznik
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, Hamilton, ON, Canada
- McMaster Institute for Research on Aging, McMaster University, Hamilton, ON, Canada
| | - Jann C Ang
- Department of Biochemistry, McMaster University, Hamilton, ON, Canada
| | - Hina Bhakta
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Angela Huynh
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Braeden Cowbrough
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, Hamilton, ON, Canada
| | - Barbara Baker
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Lauren Heessels
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Sumiya Lodhi
- Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Elizabeth Yan
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Joycelyne Ewusie
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, ON, Canada
| | - Ishac Nazy
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Jonathan Bramson
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, Hamilton, ON, Canada
| | - Matthew S Miller
- McMaster Immunology Research Centre, Hamilton, ON, Canada
- Department of Biochemistry, McMaster University, Hamilton, ON, Canada
| | - Sasha Bernatsky
- Department of Medicine, McGill University, Montreal, QC, Canada
| | - Maggie J Larché
- Department of Medicine, McMaster University, Hamilton, ON, Canada.
- McMaster Immunology Research Centre, Hamilton, ON, Canada.
| | - Dawn M E Bowdish
- Department of Medicine, McMaster University, Hamilton, ON, Canada.
- McMaster Immunology Research Centre, Hamilton, ON, Canada.
- Firestone Institute of Respiratory Health, St. Joseph's Healthcare, Hamilton, ON, Canada.
| |
Collapse
|
15
|
Andrade VM, Maricic I, Kalia R, Jachimowicz L, Bedoya O, Kulp DW, Humeau L, Smith TRF. Delineation of DNA and mRNA COVID-19 vaccine-induced immune responses in preclinical animal models. Hum Vaccin Immunother 2023; 19:2281733. [PMID: 38012018 PMCID: PMC10760386 DOI: 10.1080/21645515.2023.2281733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 11/03/2023] [Indexed: 11/29/2023] Open
Abstract
Nucleic acid vaccines are designed based on genetic sequences (DNA or mRNA) of a target antigen to be expressed in vivo to drive a host immune response. In response to the COVID-19 pandemic, mRNA and DNA vaccines based on the SARS-CoV-2 Spike antigen were developed. Surprisingly, head-to-head characterizations of the immune responses elicited by each vaccine type has not been performed to date. Here, we have employed a range of preclinical animal models including the hamster, guinea pig, rabbit, and mouse to compare and delineate the immune response raised by DNA, administered intradermally (ID) with electroporation (EP) and mRNA vaccines (BNT162b2 or mRNA-1273), administered intramuscularly (IM), expressing the SARS-CoV-2 WT spike antigen. The results revealed clear differences in the quality and magnitude of the immune response between the two vaccine platforms. The DNA vaccine immune response was characterized by strong T cell responses, while the mRNA vaccine elicited robust humoral responses. The results may assist in guiding the disease target each vaccine type may be best matched against and suggest mechanisms to further enhance the breadth of each platform's immune response.
Collapse
Affiliation(s)
| | - Igor Maricic
- Preclinical R&D, Inovio Pharmaceuticals Inc, San Diego, CA, USA
| | - Richa Kalia
- Preclinical R&D, Inovio Pharmaceuticals Inc, San Diego, CA, USA
| | | | - Olivia Bedoya
- Preclinical R&D, Inovio Pharmaceuticals Inc, San Diego, CA, USA
| | - Daniel W. Kulp
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Laurent Humeau
- Preclinical R&D, Inovio Pharmaceuticals Inc, San Diego, CA, USA
| | | |
Collapse
|
16
|
Paramithiotis E, Varaklis C, Pillet S, Shafiani S, Lancelotta MP, Steinhubl S, Sugden S, Clutter M, Montamat-Sicotte D, Chermak T, Crawford SY, Lambert BL, Mattison J, Murphy RL. Integrated antibody and cellular immunity monitoring are required for assessment of the long term protection that will be essential for effective next generation vaccine development. Front Immunol 2023; 14:1166059. [PMID: 38077383 PMCID: PMC10701527 DOI: 10.3389/fimmu.2023.1166059] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
The COVID pandemic exposed the critical role T cells play in initial immunity, the establishment and maintenance of long term protection, and of durable responsiveness against novel viral variants. A growing body of evidence indicates that adding measures of cellular immunity will fill an important knowledge gap in vaccine clinical trials, likely leading to improvements in the effectiveness of the next generation vaccines against current and emerging variants. In depth cellular immune monitoring in Phase II trials, particularly for high risk populations such as the elderly or immune compromised, should result in better understanding of the dynamics and requirements for establishing effective long term protection. Such analyses can result in cellular immunity correlates that can then be deployed in Phase III studies using appropriate, scalable technologies. Measures of cellular immunity are less established than antibodies as correlates of clinical immunity, and some misconceptions persist about cellular immune monitoring usefulness, cost, complexity, feasibility, and scalability. We outline the currently available cellular immunity assays, review their readiness for use in clinical trials, their logistical requirements, and the type of information each assay generates. The objective is to provide a reliable source of information that could be leveraged to develop a rational approach for comprehensive immune monitoring during vaccine development.
Collapse
Affiliation(s)
| | | | | | | | | | - Steve Steinhubl
- Purdue University, West Lafayette, IN, United States
- PhysIQ, Chicago, IL, United States
| | - Scott Sugden
- Medical and Scientific Affairs, Infectious Diseases, Cepheid, Sunnyvale, CA, United States
| | - Matt Clutter
- Research and Development, CellCarta, Montreal, QC, Canada
| | | | - Todd Chermak
- Regulatory and Government Affairs, CellCarta, Montreal, QC, Canada
| | - Stephanie Y. Crawford
- Department of Pharmacy Systems, Outcomes and Policy, University of Illinois Chicago, Chicago, IL, United States
| | - Bruce L. Lambert
- Department of Communication Studies, Institute for Global Health, Northwestern University, Evanston, IL, United States
| | - John Mattison
- Health Technology Advisory Board, Arsenal Capital, New York, NY, United States
| | - Robert L. Murphy
- Robert J. Havey, MD Institute for Global Health, Northwestern University, Chicago, IL, United States
| |
Collapse
|
17
|
Wang L, Nicols A, Turtle L, Richter A, Duncan CJA, Dunachie SJ, Klenerman P, Payne RP. T cell immune memory after covid-19 and vaccination. BMJ MEDICINE 2023; 2:e000468. [PMID: 38027416 PMCID: PMC10668147 DOI: 10.1136/bmjmed-2022-000468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023]
Abstract
The T cell memory response is a crucial component of adaptive immunity responsible for limiting or preventing viral reinfection. T cell memory after infection with the SARS-CoV-2 virus or vaccination is broad, and spans multiple viral proteins and epitopes, about 20 in each individual. So far the T cell memory response is long lasting and provides a high level of cross reactivity and hence resistance to viral escape by variants of the SARS-CoV-2 virus, such as the omicron variant. All current vaccine regimens tested produce robust T cell memory responses, and heterologous regimens will probably enhance protective responses through increased breadth. T cell memory could have a major role in protecting against severe covid-19 disease through rapid viral clearance and early presentation of epitopes, and the presence of cross reactive T cells might enhance this protection. T cell memory is likely to provide ongoing protection against admission to hospital and death, and the development of a pan-coronovirus vaccine might future proof against new pandemic strains.
Collapse
Affiliation(s)
- Lulu Wang
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Alex Nicols
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Lance Turtle
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- Tropical and Infectious Disease Unit, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Alex Richter
- Institute of Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Birmingham, UK
| | - Christopher JA Duncan
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
- Department of Infection and Tropical Medicine, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Susanna J Dunachie
- NDM Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University Faculty of Science, Bangkok, Thailand
| | - Paul Klenerman
- Oxford University Hospitals NHS Foundation Trust, Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, Oxfordshire, UK
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Rebecca P Payne
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
18
|
Chen Q, Chia A, Hang SK, Lim A, Koh WK, Peng Y, Gao F, Chen J, Ho Z, Wai LE, Kunasegaran K, Tan AT, Le Bert N, Loh CY, Goh YS, Renia L, Dong T, Vathsala A, Bertoletti A. Engineering immunosuppressive drug-resistant armored (IDRA) SARS-CoV-2 T cells for cell therapy. Cell Mol Immunol 2023; 20:1300-1312. [PMID: 37666955 PMCID: PMC10616128 DOI: 10.1038/s41423-023-01080-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 08/15/2023] [Indexed: 09/06/2023] Open
Abstract
Solid organ transplant (SOT) recipients receive immunosuppressive drugs (ISDs) and are susceptible to developing severe COVID-19. Here, we analyze the Spike-specific T-cell response after 3 doses of mRNA vaccine in a group of SOT patients (n = 136) treated with different ISDs. We demonstrate that a combination of a calcineurin inhibitor (CNI), mycophenolate mofetil (MMF), and prednisone (Pred) treatment regimen strongly suppressed the mRNA vaccine-induced Spike-specific cellular response. Such defects have clinical consequences because the magnitude of vaccine-induced Spike-specific T cells was directly proportional to the ability of SOT patients to rapidly clear SARS-CoV-2 after breakthrough infection. To then compensate for the T-cell defects induced by immunosuppressive treatment and to develop an alternative therapeutic strategy for SOT patients, we describe production of 6 distinct SARS-CoV-2 epitope-specific ISD-resistant T-cell receptor (TCR)-T cells engineered using the mRNA electroporation method with reactivity minimally affected by mutations occurring in Beta, Delta, Gamma, and Omicron variants. This strategy with transient expression characteristics marks an improvement in the immunotherapeutic field and provides an attractive and novel therapeutic possibility for immunosuppressed COVID-19 patients.
Collapse
Affiliation(s)
- Qi Chen
- Emerging Infectious Disease Program, Duke-NUS Medical School, Singapore, Singapore
| | - Adeline Chia
- Emerging Infectious Disease Program, Duke-NUS Medical School, Singapore, Singapore
| | - Shou Kit Hang
- Emerging Infectious Disease Program, Duke-NUS Medical School, Singapore, Singapore
| | - Amy Lim
- National University Centre for Organ Transplantation, National University Hospital, Singapore, Singapore
| | - Wee Kun Koh
- National University Centre for Organ Transplantation, National University Hospital, Singapore, Singapore
| | - Yanchun Peng
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Fei Gao
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Jili Chen
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Zack Ho
- Lion TCR Pte Ltd, Singapore, Singapore
| | - Lu-En Wai
- Lion TCR Pte Ltd, Singapore, Singapore
| | - Kamini Kunasegaran
- Emerging Infectious Disease Program, Duke-NUS Medical School, Singapore, Singapore
| | - Anthony Tanoto Tan
- Emerging Infectious Disease Program, Duke-NUS Medical School, Singapore, Singapore
| | - Nina Le Bert
- Emerging Infectious Disease Program, Duke-NUS Medical School, Singapore, Singapore
| | - Chiew Yee Loh
- A*STAR ID labs, Agency for Science, Technology and Research, Singapore, Singapore
| | - Yun Shan Goh
- A*STAR ID labs, Agency for Science, Technology and Research, Singapore, Singapore
| | - Laurent Renia
- A*STAR ID labs, Agency for Science, Technology and Research, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Tao Dong
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Anantharaman Vathsala
- National University Centre for Organ Transplantation, National University Hospital, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Antonio Bertoletti
- Emerging Infectious Disease Program, Duke-NUS Medical School, Singapore, Singapore.
- Singapore Immunology Network, A*STAR, Singapore, Singapore.
| |
Collapse
|
19
|
Neto TAP, Sidney J, Grifoni A, Sette A. Correlative CD4 and CD8 T-cell immunodominance in humans and mice: Implications for preclinical testing. Cell Mol Immunol 2023; 20:1328-1338. [PMID: 37726420 PMCID: PMC10616275 DOI: 10.1038/s41423-023-01083-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/28/2023] [Indexed: 09/21/2023] Open
Abstract
Antigen-specific T-cell recognition is restricted by Major Histocompatibility Complex (MHC) molecules, and differences between CD4 and CD8 immunogenicity in humans and animal species used in preclinical vaccine testing are yet to be fully understood. In this study, we addressed this matter by analyzing experimentally identified epitopes based on published data curated in the Immune Epitopes DataBase (IEDB) database. We first analyzed SARS-CoV-2 spike (S) and nucleoprotein (N), which are two common targets of the immune response and well studied in both human and mouse systems. We observed a weak but statistically significant correlation between human and H-2b mouse T-cell responses (CD8 S specific (r = 0.206, p = 1.37 × 10-13); CD4 S specific (r = 0.118, p = 2.63 × 10-5) and N specific (r = 0.179, p = 2.55 × 10-4)). Due to intrinsic differences in MHC molecules across species, we also investigated the association between the immunodominance of common Human Leukocyte Antigen (HLA) alleles for which HLA transgenic mice are available, namely, A*02:01, B*07:02, DRB1*01:01, and DRB1*04:01, and found higher significant correlations for both CD8 and CD4 (maximum r = 0.702, p = 1.36 × 10-31 and r = 0.594, p = 3.04-122, respectively). Our results further indicated that some regions are commonly immunogenic between humans and mice (either H-2b or HLA transgenic) but that others are human specific. Finally, we noted a significant correlation between CD8 and CD4 S- (r = 0.258, p = 7.33 × 1021) and N-specific (r = 0.369, p = 2.43 × 1014) responses, suggesting that discrete protein subregions can be simultaneously recognized by T cells. These findings were confirmed in other viral systems, providing general guidance for the use of murine models to test T-cell immunogenicity of viral antigens destined for human use.
Collapse
Affiliation(s)
- Tertuliano Alves Pereira Neto
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, 92037, USA
| | - John Sidney
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, 92037, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, 92037, USA.
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA, 92037, USA
| |
Collapse
|
20
|
Diniz MO, Maini MK, Swadling L. T cell control of SARS-CoV-2: When, which, and where? Semin Immunol 2023; 70:101828. [PMID: 37651850 DOI: 10.1016/j.smim.2023.101828] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/13/2023] [Indexed: 09/02/2023]
Abstract
Efficient immune protection against viruses such as SARS-CoV-2 requires the coordinated activity of innate immunity, B and T cells. Accumulating data point to a critical role for T cells not only in the clearance of established infection, but also for aborting viral replication independently of humoral immunity. Here we review the evidence supporting the contribution of antiviral T cells and consider which of their qualitative features favour efficient control of infection. We highlight how studies of SARS-CoV-2 and other coronaviridae in animals and humans have provided important lessons on the optimal timing (When), functionality and specificity (Which), and location (Where) of antiviral T cells. We discuss the clinical implications, particularly for the development of next-generation vaccines, and emphasise areas requiring further study.
Collapse
Affiliation(s)
- Mariana O Diniz
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London WC1E 6BT, UK
| | - Mala K Maini
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London WC1E 6BT, UK.
| | - Leo Swadling
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London WC1E 6BT, UK.
| |
Collapse
|
21
|
Neale I, Ali M, Kronsteiner B, Longet S, Abraham P, Deeks AS, Brown A, Moore SC, Stafford L, Dobson SL, Plowright M, Newman TAH, Wu MY, Crick COVID Immunity Pipeline, Carr EJ, Beale R, Otter AD, Hopkins S, Hall V, Tomic A, Payne RP, Barnes E, Richter A, Duncan CJA, Turtle L, de Silva TI, Carroll M, Lambe T, Klenerman P, Dunachie S, On behalf of the PITCH Consortium. CD4+ and CD8+ T cells and antibodies are associated with protection against Delta vaccine breakthrough infection: a nested case-control study within the PITCH study. mBio 2023; 14:e0121223. [PMID: 37655880 PMCID: PMC10653804 DOI: 10.1128/mbio.01212-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 06/26/2023] [Indexed: 09/02/2023] Open
Abstract
IMPORTANCE Defining correlates of protection against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine breakthrough infection informs vaccine policy for booster doses and future vaccine designs. Existing studies demonstrate humoral correlates of protection, but the role of T cells in protection is still unclear. In this study, we explore antibody and T cell immune responses associated with protection against Delta variant vaccine breakthrough infection in a well-characterized cohort of UK Healthcare Workers (HCWs). We demonstrate evidence to support a role for CD4+ and CD8+ T cells as well as antibodies against Delta vaccine breakthrough infection. In addition, our results suggest a potential role for cross-reactive T cells in vaccine breakthrough.
Collapse
Affiliation(s)
- Isabel Neale
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- NDM Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand
| | - Mohammad Ali
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- NDM Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand
| | - Barbara Kronsteiner
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- NDM Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Stephanie Longet
- Nuffield Department of Medicine, Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Priyanka Abraham
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- NDM Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Alexandra S. Deeks
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Anthony Brown
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Shona C. Moore
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Lizzie Stafford
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Susan L. Dobson
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Megan Plowright
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Thomas A. H. Newman
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Mary Y. Wu
- Covid Surveillance Unit, The Francis Crick Institute, London, United Kingdom
| | - Crick COVID Immunity Pipeline
- Covid Surveillance Unit, The Francis Crick Institute, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
| | | | - Rupert Beale
- The Francis Crick Institute, London, United Kingdom
- UCL Department of Renal Medicine, Royal Free Hospital, London, United Kingdom
| | | | | | | | - Adriana Tomic
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
| | - Rebecca P. Payne
- Translational and Clinical Research Institute Immunity and Inflammation Theme, Newcastle University, Newcastle, United Kingdom
| | - Eleanor Barnes
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- Translational Gastroenterology Unit, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Alex Richter
- Institute of Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Birmingham, United Kingdom
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Christopher J. A. Duncan
- Translational and Clinical Research Institute Immunity and Inflammation Theme, Newcastle University, Newcastle, United Kingdom
- Department of Infection and Tropical Medicine, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, United Kingdom
| | - Lance Turtle
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - Thushan I. de Silva
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Miles Carroll
- Nuffield Department of Medicine, Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Teresa Lambe
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, United Kingdom
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- Translational Gastroenterology Unit, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Susanna Dunachie
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- NDM Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand
- Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - On behalf of the PITCH Consortium
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- NDM Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand
- Nuffield Department of Medicine, Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
- Covid Surveillance Unit, The Francis Crick Institute, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
- UCL Department of Renal Medicine, Royal Free Hospital, London, United Kingdom
- UK Health Security Agency, Porton Down, United Kingdom
- UK Health Security Agency, London, United Kingdom
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
- Translational and Clinical Research Institute Immunity and Inflammation Theme, Newcastle University, Newcastle, United Kingdom
- Translational Gastroenterology Unit, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
- Institute of Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Birmingham, United Kingdom
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
- Department of Infection and Tropical Medicine, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, United Kingdom
- Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, United Kingdom
| |
Collapse
|
22
|
Lopez-Gomez A, Pelaez-Prestel HF, Juarez I. Approaches to evaluate the specific immune responses to SARS-CoV-2. Vaccine 2023; 41:6434-6443. [PMID: 37770298 DOI: 10.1016/j.vaccine.2023.09.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/12/2023] [Accepted: 09/19/2023] [Indexed: 09/30/2023]
Abstract
The SARS-CoV-2 pandemic has a huge impact on public health and global economy, meaning an enormous scientific, political, and social challenge. Studying how infection or vaccination triggers both cellular and humoral responses is essential to know the grade and length of protection generated in the population. Nowadays, scientists and authorities around the world are increasingly concerned about the arrival of new variants, which have a greater spread, due to the high mutation rate of this virus. The aim of this review is to summarize the different techniques available for the study of the immune responses after exposure or vaccination against SARS-CoV-2, showing their advantages and limitations, and proposing suitable combinations of different techniques to achieve extensive information in these studies. We wish that the information provided here will helps other scientists in their studies of the immune response against SARS-CoV-2 after vaccination with new vaccine candidates or infection with upcoming variants.
Collapse
Affiliation(s)
- Ana Lopez-Gomez
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Hector F Pelaez-Prestel
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.
| | - Ignacio Juarez
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
23
|
Cernera G, Gelzo M, De Placido P, Ottaviano M, Pietroluongo E, Raia M, Scalia G, Tortora M, Castaldo G, Formisano P, Palmieri G, Giuliano M. Immunocytometric analysis of patients with thymic epithelial tumors revealed that COVID-19 vaccine booster strongly enhanced the immune response. Front Immunol 2023; 14:1233056. [PMID: 37705978 PMCID: PMC10495582 DOI: 10.3389/fimmu.2023.1233056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/15/2023] [Indexed: 09/15/2023] Open
Abstract
Background Thymic epithelial tumors (TETs) are rare malignancies with heterogeneous clinical manifestations. The high frequency of autoimmune paraneoplastic disorders observed in such patients requires caution when using COVID-19 vaccines. Furthermore, TETs are often associated with severe immunodeficiency, making it difficult to predict vaccine immunization. Therefore, we aimed to evaluate immune response to COVID-19 vaccine in patients with TETs. Methods We conducted a prospective study enrolling patients who underwent the SARS-Cov-2 mRNA full vaccine cycle (two doses plus a booster after 6 months of BNT162b2). All patients were enrolled before receiving 1st vaccine dose and were followed over the vaccination cycle for up to 6 months after the booster dose to i) assess humoral and cellular responses, ii) define biomarkers predictive of effective immunization, and iii) evaluate the safety of the vaccine. Results At the end of the full vaccine cycle, 27 (61.4%) patients developed humoral and 38 (86.4%) cellular responses (IFN γ release by stimulated cells) and showed an increase in activated TH1 and TH17 cells, particularly significant after the booster dose. The number of B and T lymphocytes at baseline was predictive of humoral and cellular responses, respectively. Patients with no evidence of tumor lesions had a higher probability of achieving a humoral response than those with evidence of the disease. Furthermore, the percentage of patients with immune-related disorders (75%), particularly Good's syndrome (47.7%) and myasthenia gravis (29.5%), did not change over the entire vaccine cycle. Overall, 19 of the 44 enrolled patients (43.2%) had COVID-19 during the observation period; none required hospitalization or oxygen support, and no fatalities were observed. Conclusion SARS-Cov-2 mRNA vaccine determines the immune responses in patients with TET, particularly after the booster dose, and in patients with no evidence of tumor lesions. Preliminary analysis of B and T lymphocytes may help identify patients who have a lower probability of achieving effective humoral and cellular responses and thus may need passive immunization. The vaccine prevented severe COVID-19 infection and is safe.
Collapse
Affiliation(s)
- Gustavo Cernera
- CEINGE-Biotecnologie avanzate, scarl, Naples, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
| | - Monica Gelzo
- CEINGE-Biotecnologie avanzate, scarl, Naples, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
| | - Pietro De Placido
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, Naples, Italy
| | - Margaret Ottaviano
- Dipartimento di Melanoma, Immunoterapia Oncologica e Terapie Innovative, IRCCS Fondazione G. Pascale, Naples, Italy
- Centro Regionale di Coordinamento Tumori Rari Regione Campania (CRCTR), Naples, Italy
| | - Erica Pietroluongo
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, Naples, Italy
| | | | | | - Marianna Tortora
- Centro Regionale di Coordinamento Tumori Rari Regione Campania (CRCTR), Naples, Italy
| | - Giuseppe Castaldo
- CEINGE-Biotecnologie avanzate, scarl, Naples, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
| | - Pietro Formisano
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli Federico II, Naples, Italy
| | - Giovannella Palmieri
- Centro Regionale di Coordinamento Tumori Rari Regione Campania (CRCTR), Naples, Italy
| | - Mario Giuliano
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, Naples, Italy
- Centro Regionale di Coordinamento Tumori Rari Regione Campania (CRCTR), Naples, Italy
| |
Collapse
|
24
|
Chen Y, Mason GH, Scourfield DO, Greenshields-Watson A, Haigh TA, Sewell AK, Long HM, Gallimore AM, Rizkallah P, MacLachlan BJ, Godkin A. Structural definition of HLA class II-presented SARS-CoV-2 epitopes reveals a mechanism to escape pre-existing CD4 + T cell immunity. Cell Rep 2023; 42:112827. [PMID: 37471227 PMCID: PMC10840515 DOI: 10.1016/j.celrep.2023.112827] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/21/2023] [Accepted: 06/30/2023] [Indexed: 07/22/2023] Open
Abstract
CD4+ T cells recognize a broad range of peptide epitopes of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which contribute to immune memory and limit COVID-19 disease. We demonstrate that the immunogenicity of SARS-CoV-2 peptides, in the context of the model allotype HLA-DR1, does not correlate with their binding affinity to the HLA heterodimer. Analyzing six epitopes, some with very low binding affinity, we solve X-ray crystallographic structures of each bound to HLA-DR1. Further structural definitions reveal the precise molecular impact of viral variant mutations on epitope presentation. Omicron escaped ancestral SARS-CoV-2 immunity to two epitopes through two distinct mechanisms: (1) mutations to TCR-facing epitope positions and (2) a mechanism whereby a single amino acid substitution caused a register shift within the HLA binding groove, completely altering the peptide-HLA structure. This HLA-II-specific paradigm of immune escape highlights how CD4+ T cell memory is finely poised at the level of peptide-HLA-II presentation.
Collapse
Affiliation(s)
- Yuan Chen
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; Systems Immunity University Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Georgina H Mason
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; Systems Immunity University Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - D Oliver Scourfield
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; Systems Immunity University Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Alexander Greenshields-Watson
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; Systems Immunity University Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Tracey A Haigh
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Andrew K Sewell
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; Systems Immunity University Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Heather M Long
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Awen M Gallimore
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; Systems Immunity University Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Pierre Rizkallah
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; Systems Immunity University Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Bruce J MacLachlan
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; Systems Immunity University Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK.
| | - Andrew Godkin
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; Systems Immunity University Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; Department of Gastroenterology & Hepatology, University Hospital of Wales, Cardiff CF14 4XW, UK.
| |
Collapse
|
25
|
Grant MD, Bentley K, Fielding CA, Hatfield KM, Ings DP, Harnum D, Wang EC, Stanton RJ, Holder KA. Combined anti-S1 and anti-S2 antibodies from hybrid immunity elicit potent cross-variant ADCC against SARS-CoV-2. JCI Insight 2023; 8:e170681. [PMID: 37338994 PMCID: PMC10445686 DOI: 10.1172/jci.insight.170681] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 06/15/2023] [Indexed: 06/22/2023] Open
Abstract
Antibodies capable of neutralizing SARS-CoV-2 are well studied, but Fc receptor-dependent antibody activities that can also significantly impact the course of infection have not been studied in such depth. Since most SARS-CoV-2 vaccines induce only anti-spike antibodies, here we investigated spike-specific antibody-dependent cellular cytotoxicity (ADCC). Vaccination produced antibodies that weakly induced ADCC; however, antibodies from individuals who were infected prior to vaccination (hybrid immunity) elicited strong anti-spike ADCC. Quantitative and qualitative aspects of humoral immunity contributed to this capability, with infection skewing IgG antibody production toward S2, vaccination skewing toward S1, and hybrid immunity evoking strong responses against both domains. A combination of antibodies targeting both spike domains support strong antibody-dependent NK cell activation, with 3 regions of antibody reactivity outside the receptor-binding domain (RBD) corresponding with potent anti-spike ADCC. Consequently, ADCC induced by hybrid immunity with ancestral antigen was conserved against variants containing neutralization escape mutations in the RBD. Induction of antibodies recognizing a broad range of spike epitopes and eliciting strong and durable ADCC may partially explain why hybrid immunity provides superior protection against infection and disease compared with vaccination alone, and it demonstrates that spike-only subunit vaccines would benefit from strategies that induce combined anti-S1 and anti-S2 antibody responses.
Collapse
Affiliation(s)
- Michael D. Grant
- Immunology and Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Newfoundland, Canada
| | - Kirsten Bentley
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Ceri A. Fielding
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Keeley M. Hatfield
- Immunology and Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Newfoundland, Canada
| | - Danielle P. Ings
- Immunology and Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Newfoundland, Canada
| | - Debbie Harnum
- Eastern Health Regional Health Authority, St. John’s, Newfoundland, Canada
| | - Eddie C.Y. Wang
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Richard J. Stanton
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Kayla A. Holder
- Immunology and Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Newfoundland, Canada
| |
Collapse
|
26
|
da Silva Antunes R, Grifoni A, Frazier A, Weiskopf D, Sette A. An update on studies characterizing adaptive immune responses in SARS-CoV-2 infection and COVID-19 vaccination. Int Immunol 2023; 35:353-359. [PMID: 37148294 PMCID: PMC10406159 DOI: 10.1093/intimm/dxad014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/03/2023] [Indexed: 05/08/2023] Open
Abstract
In this brief opinion piece, we highlight our studies characterizing adaptive SARS-CoV-2 immune responses in infection and vaccination, and the ability of SARS-CoV-2-specific T cells to recognize emerging variants of concern, and the role of pre-existing cross-reactive T cells. In the context of the debate on correlates of protection, the pandemic's progression in the past 3 years underlined the need to consider how different adaptive immune responses might differentially contribute to protection from SARS-CoV-2 infection versus COVID-19 disease. Lastly, we discuss how cross-reactive T cell responses may be useful in generating a broad adaptive immunity, recognizing different variants and viral families. Considering vaccines with broadly conserved antigens could improve preparedness for future infectious disease outbreaks.
Collapse
Affiliation(s)
- Ricardo da Silva Antunes
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA 92037, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA 92037, USA
| | - April Frazier
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA 92037, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA 92037, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| |
Collapse
|
27
|
Veldhoen M, Bertoletti A. SARS-CoV-2 clearance after breakthrough infection correlates with fit and happy T cells. Immunol Cell Biol 2023; 101:587-589. [PMID: 37212205 DOI: 10.1111/imcb.12654] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Koutsakos et al. have recently published an article showing that SARS-CoV-2 breakthrough infection results in robust naïve and memory T cell activation, and the activity of CD8 T cells strongly correlates with viral clearance.
Collapse
Affiliation(s)
- Marc Veldhoen
- Instituto de Medicina Molecular, João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Antonio Bertoletti
- Emerging Infectious Diseases Program, Duke-NUS Medical School, Singapore, Singapore
- Singapore Immunology Network, A*STAR, Singapore, Singapore
| |
Collapse
|
28
|
Müller TR, Sekine T, Trubach D, Niessl J, Chen P, Bergman P, Blennow O, Hansson L, Mielke S, Nowak P, Vesterbacka J, Akber M, Olofsson A, Amaya Hernandez SP, Gao Y, Cai C, Söderdahl G, Smith CIE, Österborg A, Loré K, Sällberg Chen M, Ljungman P, Ljunggren HG, Karlsson AC, Saini SK, Aleman S, Buggert M. Additive effects of booster mRNA vaccination and SARS-CoV-2 Omicron infection on T cell immunity across immunocompromised states. Sci Transl Med 2023; 15:eadg9452. [PMID: 37437015 PMCID: PMC7615622 DOI: 10.1126/scitranslmed.adg9452] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 06/23/2023] [Indexed: 07/14/2023]
Abstract
Suboptimal immunity to SARS-CoV-2 mRNA vaccination has frequently been observed in individuals with various immunodeficiencies. Given the increased antibody evasion properties of emerging SARS-CoV-2 subvariants, it is necessary to assess whether other components of adaptive immunity generate resilient and protective responses against infection. We assessed T cell responses in 279 individuals, covering five different immunodeficiencies and healthy controls, before and after booster mRNA vaccination, as well as after Omicron infection in a subset of patients. We observed robust and persistent Omicron-reactive T cell responses that increased markedly upon booster vaccination and correlated directly with antibody titers across all patient groups. Poor vaccination responsiveness in immunocompromised or elderly individuals was effectively counteracted by the administration of additional vaccine doses. Functionally, Omicron-reactive T cell responses exhibited a pronounced cytotoxic profile and signs of longevity, characterized by CD45RA+ effector memory subpopulations with stem cell-like properties and increased proliferative capacity. Regardless of underlying immunodeficiency, booster-vaccinated and Omicron-infected individuals appeared protected against severe disease and exhibited enhanced and diversified T cell responses against conserved and Omicron-specific epitopes. Our findings indicate that T cells retain the ability to generate highly functional responses against newly emerging variants, even after repeated antigen exposure and a robust immunological imprint from ancestral SARS-CoV-2 mRNA vaccination.
Collapse
Affiliation(s)
- Thomas R. Müller
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Takuya Sekine
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Darya Trubach
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Julia Niessl
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Puran Chen
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Peter Bergman
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Laboratory Medicine, Clinical Immunology, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Ola Blennow
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Transplantation, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - Lotta Hansson
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Stephan Mielke
- Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska Comprehensive Cancer Center, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Piotr Nowak
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
- Laboratory for Molecular Infection Medicine Sweden MIMS, Umeå University, Sweden
| | - Jan Vesterbacka
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - Mira Akber
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna Olofsson
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Susana Patricia Amaya Hernandez
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Yu Gao
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Curtis Cai
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gunnar Söderdahl
- Department of Transplantation, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - C. I. Edvard Smith
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska Comprehensive Cancer Center, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Anders Österborg
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Karin Loré
- Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | - Per Ljungman
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska Comprehensive Cancer Center, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Department of Medicine Huddinge, Hematology, Karolinska Institutet, Stockholm
| | - Hans-Gustaf Ljunggren
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Annika C. Karlsson
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Laboratory, Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden
| | - Sunil Kumar Saini
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Soo Aleman
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - Marcus Buggert
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
29
|
Barnes E, Goodyear CS, Willicombe M, Gaskell C, Siebert S, I de Silva T, Murray SM, Rea D, Snowden JA, Carroll M, Pirrie S, Bowden SJ, Dunachie SJ, Richter A, Lim Z, Satsangi J, Cook G, Pope A, Hughes A, Harrison M, Lim SH, Miller P, Klenerman P, Basu N, Gilmour A, Irwin S, Meacham G, Marjot T, Dimitriadis S, Kelleher P, Prendecki M, Clarke C, Mortimer P, McIntyre S, Selby R, Meardon N, Nguyen D, Tipton T, Longet S, Laidlaw S, Orchard K, Ireland G, Thomas D, Kearns P, Kirkham A, McInnes IB. SARS-CoV-2-specific immune responses and clinical outcomes after COVID-19 vaccination in patients with immune-suppressive disease. Nat Med 2023; 29:1760-1774. [PMID: 37414897 PMCID: PMC10353927 DOI: 10.1038/s41591-023-02414-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 05/23/2023] [Indexed: 07/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immune responses and infection outcomes were evaluated in 2,686 patients with varying immune-suppressive disease states after administration of two Coronavirus Disease 2019 (COVID-19) vaccines. Overall, 255 of 2,204 (12%) patients failed to develop anti-spike antibodies, with an additional 600 of 2,204 (27%) patients generating low levels (<380 AU ml-1). Vaccine failure rates were highest in ANCA-associated vasculitis on rituximab (21/29, 72%), hemodialysis on immunosuppressive therapy (6/30, 20%) and solid organ transplant recipients (20/81, 25% and 141/458, 31%). SARS-CoV-2-specific T cell responses were detected in 513 of 580 (88%) patients, with lower T cell magnitude or proportion in hemodialysis, allogeneic hematopoietic stem cell transplantation and liver transplant recipients (versus healthy controls). Humoral responses against Omicron (BA.1) were reduced, although cross-reactive T cell responses were sustained in all participants for whom these data were available. BNT162b2 was associated with higher antibody but lower cellular responses compared to ChAdOx1 nCoV-19 vaccination. We report 474 SARS-CoV-2 infection episodes, including 48 individuals with hospitalization or death from COVID-19. Decreased magnitude of both the serological and the T cell response was associated with severe COVID-19. Overall, we identified clinical phenotypes that may benefit from targeted COVID-19 therapeutic strategies.
Collapse
Affiliation(s)
- Eleanor Barnes
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Carl S Goodyear
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Michelle Willicombe
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, London, UK
| | - Charlotte Gaskell
- Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Edgbaston, Birmingham, UK
| | - Stefan Siebert
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Thushan I de Silva
- Department of Infection, Immunity and Cardiovascular Disease, The Medical School, The University of Sheffield, Sheffield, UK
| | - Sam M Murray
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Daniel Rea
- Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Edgbaston, Birmingham, UK
| | - John A Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, UK
| | - Miles Carroll
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Sarah Pirrie
- Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Edgbaston, Birmingham, UK
| | - Sarah J Bowden
- Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Edgbaston, Birmingham, UK
| | - Susanna J Dunachie
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Alex Richter
- Clinical Immunology Service, University of Birmingham, Edgbaston, Birmingham, UK
| | - Zixiang Lim
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Jack Satsangi
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Gordon Cook
- National Institute for Health Research, Leeds MIC, University of Leeds, Leeds, UK
| | - Ann Pope
- Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Edgbaston, Birmingham, UK
| | - Ana Hughes
- Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Edgbaston, Birmingham, UK
| | - Molly Harrison
- Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Edgbaston, Birmingham, UK
| | - Sean H Lim
- Centre for Cancer Immunology, University of Southampton, Southampton, UK
| | - Paul Miller
- British Society of Blood and Marrow Transplantation and Cellular Therapy, Guy's Hospital, London, UK
| | - Paul Klenerman
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Neil Basu
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Ashley Gilmour
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Sophie Irwin
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Georgina Meacham
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Thomas Marjot
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Peter Kelleher
- Department of Infectious Diseases, Imperial College London, School of Medicine Chelsea and Westminster Hospital, London, UK
| | - Maria Prendecki
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, London, UK
| | - Candice Clarke
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, London, UK
| | - Paige Mortimer
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, London, UK
| | - Stacey McIntyre
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, London, UK
| | - Rachael Selby
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, UK
| | - Naomi Meardon
- Department of Infection, Immunity and Cardiovascular Disease, The Medical School, The University of Sheffield, Sheffield, UK
| | - Dung Nguyen
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Tom Tipton
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Stephanie Longet
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Stephen Laidlaw
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Kim Orchard
- Department of Haematology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Georgina Ireland
- UK Health Security Agency (UKHSA), Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, London, UK
| | - David Thomas
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, London, UK
| | - Pamela Kearns
- Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Edgbaston, Birmingham, UK
- National Institute for Health Research Birmingham Biomedical Research Centre, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Amanda Kirkham
- Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Edgbaston, Birmingham, UK
| | - Iain B McInnes
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
30
|
Yang H, Sun H, Brackenridge S, Zhuang X, Wing PAC, Quastel M, Walters L, Garner L, Wang B, Yao X, Felce SL, Peng Y, Moore S, Peeters BWA, Rei M, Canto Gomes J, Tomas A, Davidson A, Semple MG, Turtle LCW, Openshaw PJM, Baillie JK, Mentzer AJ, Klenerman P, Borrow P, Dong T, McKeating JA, Gillespie GM, McMichael AJ. HLA-E-restricted SARS-CoV-2-specific T cells from convalescent COVID-19 patients suppress virus replication despite HLA class Ia down-regulation. Sci Immunol 2023; 8:eabl8881. [PMID: 37390223 DOI: 10.1126/sciimmunol.abl8881] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 06/07/2023] [Indexed: 07/02/2023]
Abstract
Pathogen-specific CD8+ T cell responses restricted by the nonpolymorphic nonclassical class Ib molecule human leukocyte antigen E (HLA-E) are rarely reported in viral infections. The natural HLA-E ligand is a signal peptide derived from classical class Ia HLA molecules that interact with the NKG2/CD94 receptors to regulate natural killer cell functions, but pathogen-derived peptides can also be presented by HLA-E. Here, we describe five peptides from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that elicited HLA-E-restricted CD8+ T cell responses in convalescent patients with coronavirus disease 2019. These T cell responses were identified in the blood at frequencies similar to those reported for classical HLA-Ia-restricted anti-SARS-CoV-2 CD8+ T cells. HLA-E peptide-specific CD8+ T cell clones, which expressed diverse T cell receptors, suppressed SARS-CoV-2 replication in Calu-3 human lung epithelial cells. SARS-CoV-2 infection markedly down-regulated classical HLA class I expression in Calu-3 cells and primary reconstituted human airway epithelial cells, whereas HLA-E expression was not affected, enabling T cell recognition. Thus, HLA-E-restricted T cells could contribute to the control of SARS-CoV-2 infection alongside classical T cells.
Collapse
Affiliation(s)
- Hongbing Yang
- Centre for Immuno-Oncology, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus, Oxford, UK
- Chinese Academy of Medical Sciences Oxford Institute, Old Road Campus, Oxford, UK
| | - Hong Sun
- Centre for Immuno-Oncology, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus, Oxford, UK
- Chinese Academy of Medical Sciences Oxford Institute, Old Road Campus, Oxford, UK
- Key Laboratory of AIDS Immunology, Department of Laboratory Medicine, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Simon Brackenridge
- Centre for Immuno-Oncology, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus, Oxford, UK
| | - Xiaodong Zhuang
- Nuffield Depertment of Clinical Medicine, NDM Research Building, University of Oxford, Old Road Campus, Oxford, UK
| | - Peter A C Wing
- Chinese Academy of Medical Sciences Oxford Institute, Old Road Campus, Oxford, UK
- Nuffield Depertment of Clinical Medicine, NDM Research Building, University of Oxford, Old Road Campus, Oxford, UK
| | - Max Quastel
- Centre for Immuno-Oncology, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus, Oxford, UK
| | - Lucy Walters
- Centre for Immuno-Oncology, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus, Oxford, UK
| | - Lee Garner
- Centre for Immuno-Oncology, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus, Oxford, UK
| | - Beibei Wang
- Chinese Academy of Medical Sciences Oxford Institute, Old Road Campus, Oxford, UK
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Xuan Yao
- Chinese Academy of Medical Sciences Oxford Institute, Old Road Campus, Oxford, UK
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Suet Ling Felce
- Chinese Academy of Medical Sciences Oxford Institute, Old Road Campus, Oxford, UK
| | - Yanchun Peng
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Shona Moore
- Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Bas W A Peeters
- Centre for Immuno-Oncology, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus, Oxford, UK
| | - Margarida Rei
- Ludwig Institute for Cancer Research, University of Oxford, Old Road Campus, Oxford, UK
| | - Joao Canto Gomes
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga, Portugal
| | - Ana Tomas
- Unidada de Investigacao em Patobiologia Molecular, Instituto Portugues de Oncologia de Lisboa Francisco Gentil, EPE Lisbon, Portugal
- Chronic Diseases Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Andrew Davidson
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Malcolm G Semple
- Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- Respiratory Unit, Alder Hey Children's Hospital, Eaton Road, Liverpool L12 2AP, UK
| | - Lance C W Turtle
- Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- Tropical and Infectious Disease Unit, Liverpool University Hospitals NHS Foundation Trust (member of Liverpool Health Partners), Liverpool, UK
| | | | | | - Alexander J Mentzer
- Welcome Centre for Human Genetics, University of Oxford, Old Road Campus, Oxford, UK
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research and Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Persephone Borrow
- Centre for Immuno-Oncology, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus, Oxford, UK
| | - Tao Dong
- Chinese Academy of Medical Sciences Oxford Institute, Old Road Campus, Oxford, UK
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Jane A McKeating
- Chinese Academy of Medical Sciences Oxford Institute, Old Road Campus, Oxford, UK
- Nuffield Depertment of Clinical Medicine, NDM Research Building, University of Oxford, Old Road Campus, Oxford, UK
| | - Geraldine M Gillespie
- Centre for Immuno-Oncology, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus, Oxford, UK
| | - Andrew J McMichael
- Centre for Immuno-Oncology, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus, Oxford, UK
| |
Collapse
|
31
|
Cortese P, Amato F, Davino A, De Franchis R, Esposito S, Zollo I, Di Domenico M, Solito E, Zarrilli F, Gentile L, Cernera G, Castaldo G. The Immune Response to SARS-CoV-2 Vaccine in a Cohort of Family Pediatricians from Southern Italy. Cells 2023; 12:1447. [PMID: 37296568 PMCID: PMC10252549 DOI: 10.3390/cells12111447] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
In Italy, from January 2021, the Ministry of Health indicated a vaccination plan against COVID for frail patients and physicians based on a three-dose scheme. However, conflicting results have been reported on which biomarkers permit immunization assessment. We used several laboratory approaches (i.e., antibodies serum levels, flow cytometry analysis, and cytokines release by stimulated cells) to investigate the immune response in a cohort of 53 family pediatricians (FPs) at different times after the vaccine. We observed that the BNT162b2-mRNA vaccine induced a significant increase of specific antibodies after the third (booster) dose; however, the antibody titer was not predictive of the risk of developing the infection in the six months following the booster dose. The antigen stimulation of PBMC cells from subjects vaccinated with the third booster jab induced the increase of the activated T cells (i.e., CD4+ CD154+); the frequency of CD4+ CD154+ TNF-α+ cells, as well as the TNF-α secretion, was not modified, while we observed a trend of increase of IFN-γ secretion. Interestingly, the level of CD8+ IFN-γ+ (independently from antibody titer) was significantly increased after the third dose and predicts the risk of developing the infection in the six months following the booster jab. Such results may impact also other virus vaccinations.
Collapse
Affiliation(s)
- Paolo Cortese
- Federazione Italiana Medici Pediatri (FIMP), 80142 Naples, Italy; (P.C.); (A.D.); (R.D.F.)
| | - Felice Amato
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy; (F.A.); (I.Z.); (F.Z.); (G.C.)
- CEINGE Biotecnologie Avanzate Franco Salvatore, Scarl, 80131 Naples, Italy; (S.E.); (L.G.)
| | - Antonio Davino
- Federazione Italiana Medici Pediatri (FIMP), 80142 Naples, Italy; (P.C.); (A.D.); (R.D.F.)
| | - Raffaella De Franchis
- Federazione Italiana Medici Pediatri (FIMP), 80142 Naples, Italy; (P.C.); (A.D.); (R.D.F.)
| | - Speranza Esposito
- CEINGE Biotecnologie Avanzate Franco Salvatore, Scarl, 80131 Naples, Italy; (S.E.); (L.G.)
| | - Immacolata Zollo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy; (F.A.); (I.Z.); (F.Z.); (G.C.)
- CEINGE Biotecnologie Avanzate Franco Salvatore, Scarl, 80131 Naples, Italy; (S.E.); (L.G.)
| | - Marina Di Domenico
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania Luigi Vanvitelli, 80138 Naples, Italy;
| | - Egle Solito
- Centre for Translational Medicine and Therapeutics William Harvey Research Institute, Queen Mary Univesity, London E1 4NS, UK;
| | - Federica Zarrilli
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy; (F.A.); (I.Z.); (F.Z.); (G.C.)
- CEINGE Biotecnologie Avanzate Franco Salvatore, Scarl, 80131 Naples, Italy; (S.E.); (L.G.)
| | - Laura Gentile
- CEINGE Biotecnologie Avanzate Franco Salvatore, Scarl, 80131 Naples, Italy; (S.E.); (L.G.)
| | - Gustavo Cernera
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy; (F.A.); (I.Z.); (F.Z.); (G.C.)
- CEINGE Biotecnologie Avanzate Franco Salvatore, Scarl, 80131 Naples, Italy; (S.E.); (L.G.)
| | - Giuseppe Castaldo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy; (F.A.); (I.Z.); (F.Z.); (G.C.)
- CEINGE Biotecnologie Avanzate Franco Salvatore, Scarl, 80131 Naples, Italy; (S.E.); (L.G.)
| |
Collapse
|
32
|
Lie-Andersen O, Hübbe ML, Subramaniam K, Steen-Jensen D, Bergmann AC, Justesen D, Holmström MO, Turtle L, Justesen S, Lança T, Hansen M. Impact of peptide:HLA complex stability for the identification of SARS-CoV-2-specific CD8 +T cells. Front Immunol 2023; 14:1151659. [PMID: 37275886 PMCID: PMC10232890 DOI: 10.3389/fimmu.2023.1151659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/27/2023] [Indexed: 06/07/2023] Open
Abstract
Induction of a lasting protective immune response is dependent on presentation of epitopes to patrolling T cells through the HLA complex. While peptide:HLA (pHLA) complex affinity alone is widely exploited for epitope selection, we demonstrate that including the pHLA complex stability as a selection parameter can significantly reduce the high false discovery rate observed with predicted affinity. In this study, pHLA complex stability was measured on three common class I alleles and 1286 overlapping 9-mer peptides derived from the SARS-CoV-2 Spike protein. Peptides were pooled based on measured stability and predicted affinity. Strikingly, stability of the pHLA complex was shown to strongly select for immunogenic epitopes able to activate functional CD8+T cells. This result was observed across the three studied alleles and in both vaccinated and convalescent COVID-19 donors. Deconvolution of peptide pools showed that specific CD8+T cells recognized one or two dominant epitopes. Moreover, SARS-CoV-2 specific CD8+T cells were detected by tetramer-staining across multiple donors. In conclusion, we show that stability analysis of pHLA is a key factor for identifying immunogenic epitopes.
Collapse
Affiliation(s)
- Olivia Lie-Andersen
- Immunitrack ApS, Copenhagen, Denmark
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
- Department of Bioengineering, Technical University of Denmark, Lyngby, Denmark
| | - Mie Linder Hübbe
- Immunitrack ApS, Copenhagen, Denmark
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Krishanthi Subramaniam
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | | | | | | | - Morten Orebo Holmström
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Lance Turtle
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | | | | | - Morten Hansen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| |
Collapse
|
33
|
Murray SM, Ansari AM, Frater J, Klenerman P, Dunachie S, Barnes E, Ogbe A. The impact of pre-existing cross-reactive immunity on SARS-CoV-2 infection and vaccine responses. Nat Rev Immunol 2023; 23:304-316. [PMID: 36539527 PMCID: PMC9765363 DOI: 10.1038/s41577-022-00809-x] [Citation(s) in RCA: 87] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2022] [Indexed: 12/24/2022]
Abstract
Pre-existing cross-reactive immune responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) proteins in infection-naive subjects have been described by several studies. In particular, regions of high homology between SARS-CoV-2 and common cold coronaviruses have been highlighted as a likely source of this cross-reactivity. However, the role of such cross-reactive responses in the outcome of SARS-CoV-2 infection and vaccination is currently unclear. Here, we review evidence regarding the impact of pre-existing humoral and T cell immune responses to outcomes of SARS-CoV-2 infection and vaccination. Furthermore, we discuss the importance of conserved coronavirus epitopes for the rational design of pan-coronavirus vaccines and consider cross-reactivity of immune responses to ancestral SARS-CoV-2 and SARS-CoV-2 variants, as well as their impact on COVID-19 vaccination.
Collapse
Affiliation(s)
- Sam M Murray
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Azim M Ansari
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - John Frater
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Susanna Dunachie
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Eleanor Barnes
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK.
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK.
| | - Ane Ogbe
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
34
|
Koutsakos M, Reynaldi A, Lee WS, Nguyen J, Amarasena T, Taiaroa G, Kinsella P, Liew KC, Tran T, Kent HE, Tan HX, Rowntree LC, Nguyen THO, Thomas PG, Kedzierska K, Petersen J, Rossjohn J, Williamson DA, Khoury D, Davenport MP, Kent SJ, Wheatley AK, Juno JA. SARS-CoV-2 breakthrough infection induces rapid memory and de novo T cell responses. Immunity 2023; 56:879-892.e4. [PMID: 36958334 PMCID: PMC9970913 DOI: 10.1016/j.immuni.2023.02.017] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/26/2023] [Accepted: 02/24/2023] [Indexed: 03/24/2023]
Abstract
Although the protective role of neutralizing antibodies against COVID-19 is well established, questions remain about the relative importance of cellular immunity. Using 6 pMHC multimers in a cohort with early and frequent sampling, we define the phenotype and kinetics of recalled and primary T cell responses following Delta or Omicron breakthrough infection in previously vaccinated individuals. Recall of spike-specific CD4+ T cells was rapid, with cellular proliferation and extensive activation evident as early as 1 day post symptom onset. Similarly, spike-specific CD8+ T cells were rapidly activated but showed variable degrees of expansion. The frequency of activated SARS-CoV-2-specific CD8+ T cells at baseline and peak inversely correlated with peak SARS-CoV-2 RNA levels in nasal swabs and accelerated viral clearance. Our study demonstrates that a rapid and extensive recall of memory T cell populations occurs early after breakthrough infection and suggests that CD8+ T cells contribute to the control of viral replication in breakthrough SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Marios Koutsakos
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Wen Shi Lee
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Julie Nguyen
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Thakshila Amarasena
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - George Taiaroa
- Victorian Infectious Diseases Reference Laboratory, The Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia; Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Paul Kinsella
- Victorian Infectious Diseases Reference Laboratory, The Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Kwee Chin Liew
- Victorian Infectious Diseases Reference Laboratory, The Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Thomas Tran
- Victorian Infectious Diseases Reference Laboratory, The Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Helen E Kent
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Hyon-Xhi Tan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Louise C Rowntree
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Thi H O Nguyen
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
| | - Jan Petersen
- Infection and Immunity Program and The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia; Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, UK
| | - Deborah A Williamson
- Victorian Infectious Diseases Reference Laboratory, The Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia; Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - David Khoury
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Miles P Davenport
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jennifer A Juno
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| |
Collapse
|
35
|
Moore SC, Kronsteiner B, Longet S, Adele S, Deeks AS, Liu C, Dejnirattisai W, Reyes LS, Meardon N, Faustini S, Al-Taei S, Tipton T, Hering LM, Angyal A, Brown R, Nicols AR, Dobson SL, Supasa P, Tuekprakhon A, Cross A, Tyerman JK, Hornsby H, Grouneva I, Plowright M, Zhang P, Newman TAH, Nell JM, Abraham P, Ali M, Malone T, Neale I, Phillips E, Wilson JD, Murray SM, Zewdie M, Shields A, Horner EC, Booth LH, Stafford L, Bibi S, Wootton DG, Mentzer AJ, Conlon CP, Jeffery K, Matthews PC, Pollard AJ, Brown A, Rowland-Jones SL, Mongkolsapaya J, Payne RP, Dold C, Lambe T, Thaventhiran JED, Screaton G, Barnes E, Hopkins S, Hall V, Duncan CJA, Richter A, Carroll M, de Silva TI, Klenerman P, Dunachie S, Turtle L. Evolution of long-term vaccine-induced and hybrid immunity in healthcare workers after different COVID-19 vaccine regimens. MED 2023; 4:191-215.e9. [PMID: 36863347 PMCID: PMC9933851 DOI: 10.1016/j.medj.2023.02.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023]
Abstract
BACKGROUND Both infection and vaccination, alone or in combination, generate antibody and T cell responses against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, the maintenance of such responses-and hence protection from disease-requires careful characterization. In a large prospective study of UK healthcare workers (HCWs) (Protective Immunity from T Cells in Healthcare Workers [PITCH], within the larger SARS-CoV-2 Immunity and Reinfection Evaluation [SIREN] study), we previously observed that prior infection strongly affected subsequent cellular and humoral immunity induced after long and short dosing intervals of BNT162b2 (Pfizer/BioNTech) vaccination. METHODS Here, we report longer follow-up of 684 HCWs in this cohort over 6-9 months following two doses of BNT162b2 or AZD1222 (Oxford/AstraZeneca) vaccination and up to 6 months following a subsequent mRNA booster vaccination. FINDINGS We make three observations: first, the dynamics of humoral and cellular responses differ; binding and neutralizing antibodies declined, whereas T and memory B cell responses were maintained after the second vaccine dose. Second, vaccine boosting restored immunoglobulin (Ig) G levels; broadened neutralizing activity against variants of concern, including Omicron BA.1, BA.2, and BA.5; and boosted T cell responses above the 6-month level after dose 2. Third, prior infection maintained its impact driving larger and broader T cell responses compared with never-infected people, a feature maintained until 6 months after the third dose. CONCLUSIONS Broadly cross-reactive T cell responses are well maintained over time-especially in those with combined vaccine and infection-induced immunity ("hybrid" immunity)-and may contribute to continued protection against severe disease. FUNDING Department for Health and Social Care, Medical Research Council.
Collapse
Affiliation(s)
- Shona C Moore
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Barbara Kronsteiner
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Stephanie Longet
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sandra Adele
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Alexandra S Deeks
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Chang Liu
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Wanwisa Dejnirattisai
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Division of Emerging Infectious Disease, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Laura Silva Reyes
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Naomi Meardon
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Sian Faustini
- Institute for Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Birmingham, UK
| | - Saly Al-Taei
- Institute for Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Birmingham, UK
| | - Tom Tipton
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Luisa M Hering
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Adrienn Angyal
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Rebecca Brown
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Alexander R Nicols
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle, UK
| | - Susan L Dobson
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Piyada Supasa
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Aekkachai Tuekprakhon
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Andrew Cross
- Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Jessica K Tyerman
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle, UK
| | - Hailey Hornsby
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Irina Grouneva
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Megan Plowright
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK; Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Peijun Zhang
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Thomas A H Newman
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK; Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Jeremy M Nell
- Department of Infection and Tropical Medicine, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Priyanka Abraham
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Mohammad Ali
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Tom Malone
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Isabel Neale
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Eloise Phillips
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Joseph D Wilson
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Oxford University Medical School, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Sam M Murray
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Martha Zewdie
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Adrian Shields
- Institute for Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Birmingham, UK; University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Emily C Horner
- MRC Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Lucy H Booth
- MRC Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Lizzie Stafford
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sagida Bibi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Daniel G Wootton
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK; Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK; Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Alexander J Mentzer
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Christopher P Conlon
- Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Katie Jeffery
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Philippa C Matthews
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; The Francis Crick Institute, London, UK; Division of Infection and Immunity, University College London, London, UK; Department of Infectious Diseases, University College London Hospital NHS Foundation Trust, London, UK
| | - Andrew J Pollard
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Anthony Brown
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Sarah L Rowland-Jones
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK; Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Juthathip Mongkolsapaya
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Rebecca P Payne
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle, UK
| | - Christina Dold
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| | | | - Gavin Screaton
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Eleanor Barnes
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK; NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK; Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Susan Hopkins
- UK Health Security Agency, London, UK; Faculty of Medicine, Department of Infectious Disease, Imperial College London, London, UK; NIHR Health Protection Research Unit in Healthcare Associated Infection and Antimicrobial Resistance, University of Oxford, Oxford, UK
| | - Victoria Hall
- UK Health Security Agency, London, UK; NIHR Health Protection Research Unit in Healthcare Associated Infection and Antimicrobial Resistance, University of Oxford, Oxford, UK
| | - Christopher J A Duncan
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle, UK; Department of Infection and Tropical Medicine, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Alex Richter
- Institute for Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Birmingham, UK; University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Miles Carroll
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Thushan I de Silva
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK; Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK; NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK; Translational Gastroenterology Unit, University of Oxford, Oxford, UK.
| | - Susanna Dunachie
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand
| | - Lance Turtle
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK; Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK.
| |
Collapse
|
36
|
Walter LO, Cardoso CC, Santos-Pirath ÍM, Costa HZ, Gartner R, Werle I, Mohr ETB, da Rosa JS, Lubschinski TL, Felisberto M, Kretzer IF, Masukawa II, Vanny PDA, Luiz MC, de Moraes ACR, Dalmarco EM, Santos-Silva MC. T cell maturation is significantly affected by SARS-CoV-2 infection. Immunology 2023. [PMID: 36855300 DOI: 10.1111/imm.13635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a respiratory tract infection caused by the new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). An adequate T cell response is essential not only for fighting disease but also for the creation of immune memory. Thus, the present study aims to evaluate the T cells of patients with moderate, severe and critical COVID-19 not only at the time of illness but also 2 months after diagnosis to observe whether changes in this compartment persist. In this study, 166 COVID-19 patients were stratified into moderate/severe and critical disease categories. The maturation and activation of T cells were evaluated through flow cytometry. In addition, Treg cells were analysed. Until 15 days after diagnosis, patients presented a reduction in absolute and relative T lymphocyte counts. After 2 months, in moderate/severe patients, the counts returned to a similar level as that of the control group. In convalescent patients who had a critical illness, absolute T lymphocyte values increased considerably. Patients with active disease did not show differentiation of T cells. Nonetheless, after 2 months, patients with critical COVID-19 showed a significant increase in CD4+ EMRA (CD45RA+ effector memory) T lymphocytes. Furthermore, COVID-19 patients showed delayed T cell activation and reduced CD8+ suppressor T cells even 2 months after diagnosis. A reduction in CD4+ Treg cells was also observed, and their numbers returned to a similar level as that of healthy controls in convalescent patients. The results demonstrate that COVID-19 patients have a delayed activation and differentiation of T cells. In addition, these patients have a great reduction of T cells with a suppressor phenotype.
Collapse
Affiliation(s)
- Laura Otto Walter
- Postgraduate Program in Pharmacy, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Chandra Chiappin Cardoso
- Division of Clinical Analysis, Flow Cytometry Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil
| | - Íris Mattos Santos-Pirath
- Division of Clinical Analysis, Flow Cytometry Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil
| | - Heloisa Zorzi Costa
- Division of Clinical Analysis, Flow Cytometry Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil
| | - Rafaela Gartner
- Clinical Analysis Department, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Isabel Werle
- Clinical Analysis Department, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, Brazil
| | | | - Julia Salvan da Rosa
- Postgraduate Program in Pharmacy, Federal University of Santa Catarina, Florianópolis, Brazil
| | | | - Mariano Felisberto
- Postgraduate Program in Pharmacy, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Iara Fabricia Kretzer
- Clinical Analysis Department, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Ivete Ioshiko Masukawa
- Infectious Disease Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil.,Infectious Disease Service, Nereu Ramos Hospital, State Health Department, Florianópolis, Brazil
| | - Patrícia de Almeida Vanny
- Infectious Disease Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil
| | - Magali Chaves Luiz
- Infectious Disease Service, Nereu Ramos Hospital, State Health Department, Florianópolis, Brazil
| | - Ana Carolina Rabello de Moraes
- Postgraduate Program in Pharmacy, Federal University of Santa Catarina, Florianópolis, Brazil.,Division of Clinical Analysis, Flow Cytometry Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil
| | - Eduardo Monguilhott Dalmarco
- Postgraduate Program in Pharmacy, Federal University of Santa Catarina, Florianópolis, Brazil.,Division of Clinical Analysis, Flow Cytometry Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil
| | - Maria Cláudia Santos-Silva
- Postgraduate Program in Pharmacy, Federal University of Santa Catarina, Florianópolis, Brazil.,Division of Clinical Analysis, Flow Cytometry Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil.,Clinical Analysis Department, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
37
|
Swadling L, Maini MK. Can T Cells Abort SARS-CoV-2 and Other Viral Infections? Int J Mol Sci 2023; 24:4371. [PMID: 36901802 PMCID: PMC10002440 DOI: 10.3390/ijms24054371] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Despite the highly infectious nature of the SARS-CoV-2 virus, it is clear that some individuals with potential exposure, or even experimental challenge with the virus, resist developing a detectable infection. While a proportion of seronegative individuals will have completely avoided exposure to the virus, a growing body of evidence suggests a subset of individuals are exposed, but mediate rapid viral clearance before the infection is detected by PCR or seroconversion. This type of "abortive" infection likely represents a dead-end in transmission and precludes the possibility for development of disease. It is, therefore, a desirable outcome on exposure and a setting in which highly effective immunity can be studied. Here, we describe how early sampling of a new pandemic virus using sensitive immunoassays and a novel transcriptomic signature can identify abortive infections. Despite the challenges in identifying abortive infections, we highlight diverse lines of evidence supporting their occurrence. In particular, expansion of virus-specific T cells in seronegative individuals suggests abortive infections occur not only after exposure to SARS-CoV-2, but for other coronaviridae, and diverse viral infections of global health importance (e.g., HIV, HCV, HBV). We discuss unanswered questions related to abortive infection, such as: 'Are we just missing antibodies? Are T cells an epiphenomenon? What is the influence of the dose of viral inoculum?' Finally, we argue for a refinement of the current paradigm that T cells are only involved in clearing established infection; instead, we emphasise the importance of considering their role in terminating early viral replication by studying abortive infections.
Collapse
Affiliation(s)
- Leo Swadling
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London WC1E 6BT, UK
| | - Mala K. Maini
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London WC1E 6BT, UK
| |
Collapse
|
38
|
Peng SS, Shao XB, Gu MX, Zhang GS, Gu C, Nian Y, Jia Y, Han Y, Liu XQ, Sun LB. Catalytically Stable Potassium Single-Atom Solid Superbases. Angew Chem Int Ed Engl 2022; 61:e202215157. [PMID: 36333269 DOI: 10.1002/anie.202215157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Indexed: 11/06/2022]
Abstract
Solid superbases can catalyze diverse reactions under mild conditions, while they suffer from aggregation of basic sites and poor stability during recycling. Here we report a new generation of solid superbases derived from K single atoms (SAs) prepared by a tandem redox strategy. The initial redox reaction takes place between base precursor KNO3 and graphene support, producing K2 O at 400 °C. Further increasing the temperature to 800 °C, the graphene reduces K2 O to K anchored by its vacancies, leading to the generation of K SAs (denoted as K1 /G). The source of basicity in the K1 /G is K SAs, and neighboring single atoms (NSAs) possess superbasicity, which is different from conventional basicity originated from oxygen and nitrogen atoms. Due to the superbasicity as well as high dispersion and anchoring of basic sites, the K1 /G shows excellent catalytic activity and stability in transesterification reaction, which is much superior to the reported catalysts.
Collapse
Affiliation(s)
- Song-Song Peng
- State Key Laboratory of Materials-Oriented Chemical Engineering, Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), College of Chemical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Xiang-Bin Shao
- State Key Laboratory of Materials-Oriented Chemical Engineering, Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), College of Chemical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Meng-Xuan Gu
- State Key Laboratory of Materials-Oriented Chemical Engineering, Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), College of Chemical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Guo-Song Zhang
- State Key Laboratory of Materials-Oriented Chemical Engineering, Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), College of Chemical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Chen Gu
- State Key Laboratory of Materials-Oriented Chemical Engineering, Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), College of Chemical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Yao Nian
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Yiming Jia
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - You Han
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China.,Haihe Laboratory of Sustainable Chemical Transformations, Tianjin, 300192, China
| | - Xiao-Qin Liu
- State Key Laboratory of Materials-Oriented Chemical Engineering, Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), College of Chemical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Lin-Bing Sun
- State Key Laboratory of Materials-Oriented Chemical Engineering, Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), College of Chemical Engineering, Nanjing Tech University, Nanjing, 211816, China
| |
Collapse
|
39
|
Antibody and T cell responses against wild-type and Omicron SARS-CoV-2 after third-dose BNT162b2 in adolescents. Signal Transduct Target Ther 2022; 7:397. [PMID: 36517469 PMCID: PMC9748396 DOI: 10.1038/s41392-022-01282-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 11/30/2022] [Accepted: 12/04/2022] [Indexed: 12/15/2022] Open
Abstract
The high effectiveness of the third dose of BNT162b2 in healthy adolescents against Omicron BA.1 has been reported in some studies, but immune responses conferring this protection are not yet elucidated. In this analysis, our study (NCT04800133) aims to evaluate the humoral and cellular responses against wild-type and Omicron (BA.1, BA.2 and/or BA.5) SARS-CoV-2 before and after a third dose of BNT162b2 in healthy adolescents. At 5 months after 2 doses, S IgG, S IgG Fc receptor-binding, and neutralising antibody responses waned significantly, yet neutralising antibodies remained detectable in all tested adolescents and S IgG avidity increased from 1 month after 2 doses. The antibody responses and S-specific IFN-γ+ and IL-2+ CD8+ T cell responses were significantly boosted in healthy adolescents after a homologous third dose of BNT162b2. Compared to adults, humoral responses for the third dose were non-inferior or superior in adolescents. The S-specific IFN-γ+ and IL-2+ CD4+ and CD8+ T cell responses in adolescents and adults were comparable or non-inferior. Interestingly, after 3 doses, adolescents had preserved S IgG, S IgG avidity, S IgG FcγRIIIa-binding, against Omicron BA.2, as well as preserved cellular responses against BA.1 S and moderate neutralisation levels against BA.1, BA.2 and BA.5. Sera from 100 and 96% of adolescents tested at 1 and 5 months after two doses could also neutralise BA.1. Our study found high antibody and T cell responses, including potent cross-variant reactivity, after three doses of BNT162b2 vaccine in adolescents in its current formulation, suggesting that current vaccines can be protective against symptomatic Omicron disease.
Collapse
|