1
|
Jagne YJ, Jobe D, Darboe A, Danso M, Barratt N, Gomez M, Wenlock R, Jarju S, Sylva EL, Touray AF, Toure F, Kumado M, Saso A, Zafred D, Nicklin M, Sayers J, Hornsby H, Lindsey B, Sesay AK, Temperton N, Kucharski A, Hodgson D, de Silva T, Kampmann B. Compartmentalised mucosal and blood immunity to SARS-CoV-2 is associated with high seroprevalence before the Delta wave in Africa. COMMUNICATIONS MEDICINE 2025; 5:178. [PMID: 40379979 DOI: 10.1038/s43856-025-00902-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 05/09/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND The reported number of SARS-CoV-2 cases and deaths are lower in Africa compared to many high-income countries. However, in African cohorts, detailed characterisation of SARS-CoV-2 mucosal and T cell immunity are limited. We assessed the SARS-CoV-2-specific immune landscape in The Gambia during the presence of the pre-Delta variant in July 2021. METHODS A cross-sectional assessment of SARS-CoV-2 immunity in 349 unvaccinated individuals from 52 Gambian households was performed between March-June 2021. SARS-CoV-2 spike (S) and nucleocapsid (N) specific binding antibodies were measured by ELISA, variant-specific serum neutralizing-antibodies (NAb) by viral pseudotype assays and nasal fluid IgA by mesoscale discovery assay. SARS-CoV-2 T-cell responses were evaluated using ELISpot assay. RESULTS We show that adjusted anti-Spike antibody seroprevalence is 56.7% (95% confidence interval (CI) 49.0-64.0), with lower rates in children <5 years (26.2%, 13.9-43.8) and 5-17 years (46.4%, 36.2-56.7) compared to adults 18-49 years (78.4%, 68.8-85.8). Among spike-seropositive individuals, NAb titres are highest against Alpha variant (median IC50 110), with 27% showing pre-existing Delta variant titres >1:50. T-cell responses are higher in spike-seropositive individuals, although 34% of spike-seronegative individuals show responses to at least one antigen pool. We observe strong correlations within SARS-CoV-2 T-cell, mucosal IgA, and serum NAb responses. CONCLUSIONS High SARS-CoV-2 seroprevalence in The-Gambia induce mucosal and blood immunity, reducing Delta and Omicron impact. Children are relatively protected from infection. T-cell responses in seronegative individuals may indicate either pre-pandemic cross-reactivity or individuals with a T-cell dominated response to SARS-CoV-2 infection with absent or poor humoral responses.
Collapse
Affiliation(s)
- Ya Jankey Jagne
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia.
| | - Dawda Jobe
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Alansana Darboe
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Madikoi Danso
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Natalie Barratt
- Division of Clinical Medicine, School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
- The Florey Institute of Infection, The University of Sheffield, Sheffield, UK
| | - Marie Gomez
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Rhys Wenlock
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Sheikh Jarju
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Ellen Lena Sylva
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Aji Fatou Touray
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Fatoumata Toure
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Michelle Kumado
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Anja Saso
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Domen Zafred
- Division of Clinical Medicine, School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
| | - Martin Nicklin
- Division of Clinical Medicine, School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
| | - Jon Sayers
- Division of Clinical Medicine, School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
| | - Hailey Hornsby
- Division of Clinical Medicine, School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
- The Florey Institute of Infection, The University of Sheffield, Sheffield, UK
| | - Benjamin Lindsey
- Division of Clinical Medicine, School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
- The Florey Institute of Infection, The University of Sheffield, Sheffield, UK
| | - Abdul Karim Sesay
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Canterbury, UK
| | - Adam Kucharski
- Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London, UK
| | - David Hodgson
- Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London, UK
| | - Thushan de Silva
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
- Division of Clinical Medicine, School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
- The Florey Institute of Infection, The University of Sheffield, Sheffield, UK
| | - Beate Kampmann
- Vaccines and Immunity Theme, Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia.
- Charité Centre for Global Health; Charité Universitätsmedizin, Berlin, Germany.
| |
Collapse
|
2
|
Hodgson D, Liu Y, Carolan L, Mahanty S, Subbarao K, Sullivan SG, Fox A, Kucharski A. Memory B cell proliferation drives differences in neutralising responses between ChAdOx1 and BNT162b2 SARS-CoV-2 vaccines. Front Immunol 2025; 16:1487066. [PMID: 40196126 PMCID: PMC11974255 DOI: 10.3389/fimmu.2025.1487066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Vaccination against COVID-19 has been pivotal in reducing the global burden of the disease. However, Phase III trial results and observational studies underscore differences in efficacy across vaccine technologies and dosing regimens. Notably, mRNA vaccines have exhibited superior effectiveness compared to Adenovirus (AdV) vaccines, especially with extended dosing intervals. Methods Using in-host mechanistic modelling, this study elucidates these variations and unravels the biological mechanisms shaping the immune responses at the cellular level. We used data on the change in memory B cells, plasmablasts, and antibody titres after the second dose of a COVID-19 vaccine for Australian healthcare workers. Alongside this dataset, we constructed a kinetic model of humoral immunity which jointly captured the dynamics of multiple immune markers, and integrated hierarchical effects into this kinetics model, including age, dosing schedule, and vaccine type. Results Our analysis estimated that mRNA vaccines induced 2.1 times higher memory B cell proliferation than AdV vaccines after adjusting for age, interval between doses and priming dose. Additionally, extending the duration between the second vaccine dose and priming dose beyond 28 days boosted neutralising antibody production per plasmablast concentration by 30%. We also found that antibody responses after the second dose were more persistent when mRNA vaccines were used over AdV vaccines and for longer dosing regimens. Discussion Reconstructing in-host kinetics in response to vaccination could help optimise vaccine dosing regimens, improve vaccine efficacy in different population groups, and inform the design of future vaccines for enhanced protection against emerging pathogens.
Collapse
Affiliation(s)
- David Hodgson
- Centre of Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Yi Liu
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Louise Carolan
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Siddhartha Mahanty
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Kanta Subbarao
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Sheena G. Sullivan
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- School of Clinical Sciences, Monash University, Melbourne, VIC, Australia
| | - Annette Fox
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Adam Kucharski
- Centre of Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
3
|
Fahnøe U, Feng S, Underwood AP, Jacobsen K, Ameri A, Blicher TH, Sølund CS, Rosenberg BR, Brix L, Weis N, Bukh J. T cell receptor usage and epitope specificity amongst CD8 + and CD4 + SARS-CoV-2-specific T cells. Front Immunol 2025; 16:1510436. [PMID: 40092978 PMCID: PMC11906682 DOI: 10.3389/fimmu.2025.1510436] [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: 10/12/2024] [Accepted: 01/02/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has highlighted the critical importance of understanding protective long-lasting immune responses. This study investigates the epitope specificity, T cell receptor (TCR) usage, and phenotypic changes in SARS-CoV-2-specfic CD8+ and CD4+ T cells over time in convalescent individuals with COVID-19. Methods Peripheral blood mononuclear cells (PBMCs) were collected from 28 unvaccinated individuals with primary SARS-CoV-2 infection (6 identified as the D614G variant, clade 20C) and analyzed up to 12 months post-symptom onset. Antigen-specific CD8+ and CD4+ T cells were analyzed using flow cytometry and single-cell RNA sequencing (scRNAseq) using specific dextramer and antibody reagents. TCR clonotypes and activation markers were characterized to explore T cell dynamics. Results SARS-CoV-2-specific CD8+ T cells exhibited waning frequencies long-term, transitioning from memory-like to a naïve-like state. scRNAseq revealed specificity against both spike and non-spike antigens with increased CD95 and CD127 expression over time, indicating that naïve-like T cells may represent stem cell memory T cells, which are multipotent and self-renewing, likely important for long-lived immunity. TCR clonal expansion was observed mainly in memory T cells, with overlapping TCR beta chain (TRB)-complementary determining region 3 (CDR3) sequences between participants, suggesting shared public TCR epitope-specific repertoires against SARS-CoV-2. Further, unique spike-specific CD4+ T cells with high CD95 and CD127 expression were identified, which may play a crucial role in long-term protection. Discussion This study highlights epitope-specificity heterogeneity, with some immunodominant responses, and suggests a potential role for long-lived SARS-CoV-2-specific T cell immunity. Shared TCR repertoires offers insights into cross-reactive and protective T cell clones, providing valuable information for optimizing vaccine strategies against emerging SARS-CoV-2 variants. The findings underscore the critical role of cellular immunity in long-term protection against SARS-CoV-2 and emphasizes the importance of understanding T cell dynamics.
Collapse
Affiliation(s)
- Ulrik Fahnøe
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Shan Feng
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Alexander P. Underwood
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | | | | | | | - Christina S. Sølund
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Brad R. Rosenberg
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Nina Weis
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Bukh
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| |
Collapse
|
4
|
Sidney J, Kim AR, de Vries RD, Peters B, Meade PS, Krammer F, Grifoni A, Sette A. Targets of influenza human T-cell response are mostly conserved in H5N1. mBio 2025; 16:e0347924. [PMID: 39714185 PMCID: PMC11796400 DOI: 10.1128/mbio.03479-24] [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: 11/19/2024] [Accepted: 12/05/2024] [Indexed: 12/24/2024] Open
Abstract
Frequent recent spillovers of subtype H5N1 clade 2.3.4.4b highly pathogenic avian influenza (HPAI) virus into poultry and mammals, especially dairy cattle, including several human cases, increased concerns over a possible future pandemic. Here, we performed an analysis of epitope data curated in the Immune Epitope Database (IEDB). We found that the patterns of immunodominance of seasonal influenza viruses circulating in humans and H5N1 are similar. We further conclude that a significant fraction of the T-cell epitopes is conserved at a level associated with cross-reactivity between avian and seasonal sequences, and we further experimentally demonstrate extensive cross-reactivity in the most dominant T-cell epitopes curated in the IEDB. Based on these observations, and the overall similarity of the neuraminidase (NA) N1 subtype encoded in both HPAI and seasonal H1N1 influenza virus as well as cross-reactive group 1 HA stalk-reactive antibodies, we expect that a degree of pre-existing immunity is present in the general human population that could blunt the severity of human H5N1 infections.IMPORTANCEInfluenza A viruses (IAVs) cause pandemics that can result in millions of deaths. The highly pathogenic avian influenza (HPAI) virus of the H5N1 subtype is presently among the top viruses of pandemic concern, according to the WHO and the National Institute of Allergy and Infectious Diseases (NIAID). Previous exposure by infection and/or vaccination to a given IAV subtype or clade influences immune responses to a different subtype or clade. Analysis of human CD4 and CD8 T-cell epitope conservation between HPAI H5N1 and seasonal IAV sequences revealed levels of identity and conservation conducive to T cell cross-reactivity, suggesting that pre-existing T cell immune memory should, to a large extent, cross-recognize avian influenza viruses. This observation was experimentally verified by testing responses from human T cells to non-avian IAV and their HPAI H5N1 counterparts. Accordingly, should a more widespread HPAI H5N1 outbreak occur, we hypothesize that cross-reactive T-cell responses might be able to limit disease severity.
Collapse
Affiliation(s)
- John Sidney
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, California, USA
| | - A-Reum Kim
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, California, USA
| | - Rory D. de Vries
- Department of Viroscience, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Bjoern Peters
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, California, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, California, USA
| | - Philip S. Meade
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| | - Alba Grifoni
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, California, USA
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, California, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, California, USA
| |
Collapse
|
5
|
Huang K, Li N, Li Y, Zhu J, Fan Q, Yang J, Gao Y, Liu Y, Gao S, Zhao P, Wei K, Deng C, Zuo C, Sun Z. Circular mRNA Vaccine against SARS-COV-2 Variants Enabled by Degradable Lipid Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2025; 17:4699-4710. [PMID: 39789795 DOI: 10.1021/acsami.4c20770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
The emergence of mRNA vaccines offers great promise and a potent platform in combating various diseases, notably COVID-19. Nevertheless, challenges such as inherent instability and potential side effects of current delivery systems underscore the critical need for the advancement of stable, safe, and efficacious mRNA vaccines. In this study, a robust mRNA vaccine (cmRNA-1130) eliciting potent immune activation has been developed from a biodegradable lipid with eight ester bonds in the branched tail (AX4) and synthetic circular mRNA (cmRNA) encoding the trimeric Delta receptor binding domain of the SARS-CoV-2 spike protein. Notably, the cmRNA-1130 vaccine exhibits outstanding stability, remaining effective after six months of storage at 4 °C and multiple freeze-thaw cycles. In comparison with the commercial MC3 lipid, the nanoparticles formed from the degradable AX4 lipid revealed a much faster metabolic rate from the liver and spleen, affording negligible impairment to the hepatorenal function. Following intramuscular administration, cmRNA-1130 generates robust and sustained neutralizing antibodies and induces the activation of Delta RBD-specific CD4+ and CD8+ T effector memory cells (TEM) and Th1-biased T cells in mice. Featured with potent immune activation, high stability, and decent safety, vaccines formed from cmRNA and AX4 hold a huge clinical potential for the prophylaxis and treatment of different diseases.
Collapse
Affiliation(s)
- Ke Huang
- Department of Chemical and Biological Engineering, Monash University, Clayton 3800, Victoria, Australia
| | - Na Li
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Yingwen Li
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Jiafeng Zhu
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Qianyi Fan
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Jiali Yang
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Yinjia Gao
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Yuping Liu
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Shufeng Gao
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Peng Zhao
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Ke Wei
- Hunan University of Chinese Medicine, Changsha 410208, China
| | - Chao Deng
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Materials, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Chijian Zuo
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Zhenhua Sun
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| |
Collapse
|
6
|
da Silva Antunes R, Fajardo-Rosas V, Yu ED, Gálvez RI, Abawi A, Alexandar Escarrega E, Martínez-Pérez A, Johansson E, Goodwin B, Frazier A, Dan JM, Crotty S, Seumois G, Weiskopf D, Vijayanand P, Sette A. Evolution of SARS-CoV-2 T cell responses as a function of multiple COVID-19 boosters. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.08.631842. [PMID: 39829792 PMCID: PMC11741356 DOI: 10.1101/2025.01.08.631842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The long-term effects of repeated COVID-19 vaccinations on adaptive immunity remain incompletely understood. Here, we conducted a comprehensive three-year longitudinal study examining T cell and antibody responses in 78 vaccinated individuals without reported symptomatic infections. We observed distinct dynamics in Spike-specific humoral and cellular immune responses across multiple vaccine doses. While antibody titers incrementally increased and stabilized with each booster, T cell responses rapidly plateaued, maintaining remarkable stability across CD4+ and CD8+ subsets. Notably, approximately 30% of participants showed CD4+ T cell reactivity to non-Spike antigens, consistent with asymptomatic infections. Single-cell RNA sequencing revealed a diverse landscape of Spike-specific T cell phenotypes, with no evidence of increased exhaustion or significant functional impairment. However, qualitative changes were observed in individuals with evidence of asymptomatic infection, exhibiting unique immunological characteristics, including increased frequencies of Th17-like CD4+ T cells and GZMKhi/IFNR CD8+ T cell subsets. Remarkably, repeated vaccinations in this group were associated with a progressive increase in regulatory T cells, potentially indicating a balanced immune response that may mitigate immunopathology. By regularly stimulating T cell memory, boosters contribute to a stable and enhanced immune response, which may provide better protection against symptomatic infections.
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
- These authors contributed equally
| | - Vicente Fajardo-Rosas
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Bioinformatics and Systems Biology Graduate Program; University of California, San Diego, La Jolla, CA 92093, USA
- These authors contributed equally
| | - Esther Dawen Yu
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Rosa Isela Gálvez
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Adam Abawi
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - E. Alexandar Escarrega
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Amparo Martínez-Pérez
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Emil Johansson
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Benjamin Goodwin
- 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
| | - Jennifer M. Dan
- 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 92037, USA
| | - Shane Crotty
- 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 92037, USA
| | - Grégory Seumois
- 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
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Pandurangan Vijayanand
- 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 92037, USA
- Senior authorship
| | - 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 92037, USA
- Senior authorship
- Lead contact
| |
Collapse
|
7
|
Li Y, Lin Y, Yi Y, Zhu N, Cui X, Li X. COVID-19 Vaccination and Transient Increase in CD4/CD8 Cell Counts in People with HIV: Evidence from China. Vaccines (Basel) 2024; 12:1365. [PMID: 39772028 PMCID: PMC11680300 DOI: 10.3390/vaccines12121365] [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: 10/17/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
Objectives: Accumulating evidence has confirmed the efficacy and safety of COVID-19 vaccines against SARS-CoV-2 infection. However, the effect of COVID-19 vaccination on immuno-virological parameters in people with HIV (PWH) is uncertain. Methods: A total of 372 PWH treated at Beijing Ditan Hospital were included. Unvaccinated PWH were matched 1:3 with vaccinated PWH using a propensity score matching algorithm. Differences in immuno-virological markers between the matched groups were analyzed. The Wilcoxon signed rank test was used to test for changes in CD4 and CD8 counts and HIV viral load over two months around vaccination. In addition, we investigated the long-term changes in HIV-related markers in different vaccination dose groups and in the entire vaccinated population. Results: Vaccinated PWH had a higher CD4/CD8 ratio (0.64 (0.49, 0.78) vs. 0.80 (0.56, 1.03), p = 0.037) than unvaccinated PWH within a two-month window after the third dose. There were 337 PWH who received COVID-19 vaccination, and 73.9% (n = 249) received three doses of vaccine. We observed a transient increase in CD4 count and CD4/CD8 ratio within a two-month window after vaccination, especially after the second dose (CD4 count: 583.5 (428.5, 706.8) vs. 618.0 (452.0, 744.0), p = 0.018; CD4/CD8 ratio: 0.70 (0.50, 0.91) vs. 0.71 (0.53, 0.96), p < 0.001)) and the third dose (CD4 count: 575.5 (435.5, 717.0) vs. 577.5 (440.8, 754.8), p = 0.001; CD4/CD8 ratio: 0.70 (0.52, 0.93) vs. 0.79 (0.53, 1.00), p < 0.001)). Recent CD4 counts and CD4/CD8 ratios were lower than after COVID-19 but remained higher than before COVID-19 in vaccinated PWH. In addition, COVID-19 vaccination had no negative effect on HIV viral load. Conclusions: A transient increase in CD4 count and CD4/CD8 ratio was observed after COVID-19 vaccination. However, the enhanced cellular immune response induced by vaccination may diminish over time and return to normal levels. There is no adverse effect of vaccination on HIV viral load.
Collapse
Affiliation(s)
- Yanyan Li
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; (Y.L.); (N.Z.); (X.C.)
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Yingying Lin
- Center of Integrative Medicine, Peking University Ditan Teaching Hospital, Beijing 100015, China;
| | - Yunyun Yi
- Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, PLA General Hospital, Beijing 100853, China;
| | - Na Zhu
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; (Y.L.); (N.Z.); (X.C.)
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xinyu Cui
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; (Y.L.); (N.Z.); (X.C.)
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xin Li
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; (Y.L.); (N.Z.); (X.C.)
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
- Center of Integrative Medicine, Peking University Ditan Teaching Hospital, Beijing 100015, China;
| |
Collapse
|
8
|
Mvula M, Mtonga F, Mandolo J, Jowati C, Kalirani A, Chigamba P, Lisimba E, Mitole N, Chibwana MG, Jambo KC. Longevity of hybrid immunity against SARS-CoV-2 in adults vaccinated with an adenovirus-based COVID-19 vaccine. BMC Infect Dis 2024; 24:959. [PMID: 39266969 PMCID: PMC11391831 DOI: 10.1186/s12879-024-09891-z] [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: 05/03/2024] [Accepted: 09/05/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Hybrid immunity provides better protection against COVID-19 than vaccination or prior natural infection alone. It induces high magnitude and broadly cross-reactive neutralising anti-Spike IgG antibodies. However, it is not clear how long these potent antibodies last, especially in the context of adenovirus-based COVID-19 vaccines. METHODS We conducted a longitudinal cohort study and enrolled 20 adults who had received an adenovirus-based COVID-19 vaccine before a laboratory-confirmed SARS-CoV-2 infection. We followed up the study participants for 390 days post the initial breakthrough infection. We assessed the longevity and cross-reactive breadth of serum antibodies against SARS-CoV-2 variants of concern (VOCs), including Omicron. RESULTS The binding anti-Spike IgG antibodies remained within the reported putative levels for at least 360 days and were cross-neutralising against Beta, Gamma, Delta, and Omicron. During the follow up period, a median of one SARS-CoV-2 re-infection event was observed across the cohort, but none resulted in severe COVID-19. Moreover, the re-exposure events were associated with augmented anti-Spike and anti-RBD IgG antibody titres. CONCLUSIONS This study confirms that hybrid immunity provides durable broadly cross-reactive antibody immunity against SARS-CoV-2 variants of concern for at least a year (360 days), and that it is further augment by SARS-CoV-2 re-exposure.
Collapse
Affiliation(s)
- Memory Mvula
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - Fatima Mtonga
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - Jonathan Mandolo
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
- Liverpool School of Tropical Medicine, Liverpool, UK
| | - Chisomo Jowati
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - Alice Kalirani
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | | | - Edwin Lisimba
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - Ndaona Mitole
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - Marah G Chibwana
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
- University of Oxford, Oxford, England, UK
| | - Kondwani C Jambo
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi.
- Liverpool School of Tropical Medicine, Liverpool, UK.
| |
Collapse
|
9
|
Wee EGT, Kempster S, Ferguson D, Hall J, Ham C, Morris S, Crook A, Gilbert SC, Korber B, Almond N, Hanke T. Design, Immunogenicity and Preclinical Efficacy of the ChAdOx1.COVconsv12 Pan-Sarbecovirus T-Cell Vaccine. Vaccines (Basel) 2024; 12:965. [PMID: 39339997 PMCID: PMC11436245 DOI: 10.3390/vaccines12090965] [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: 07/29/2024] [Revised: 08/22/2024] [Accepted: 08/24/2024] [Indexed: 09/30/2024] Open
Abstract
During the COVID-19 pandemic, antibody-based vaccines targeting the SARS-CoV-2 spike glycoprotein were the focus for development because neutralizing antibodies were associated with protection against the SARS-CoV-2 infection pre-clinically and in humans. While deploying these spike-based vaccines saved millions of lives worldwide, it has become clear that the immunological mechanisms of protection against severe disease are multifaceted and involve non-neutralizing antibody components. Here, we describe a novel pan-sarbecovirus T-cell vaccine, ChAdOx1.COVconsv12, designed to complement and broaden the protection of spike vaccines. The vaccine immunogen COVconsv12 employs the two regions in the viral proteome most conserved among sarbecoviruses, which are delivered by replication-deficient vector ChAdOx1. It directs T cells towards epitopes shared among sarbecoviruses including evolving SARS-CoV-2 variants. Here, we show that ChAdOx1.COVconsv12 induced broad T-cell responses in the BALB/c and C57BL/6 mice. In the Syrian hamster challenge model, ChAdOx1.COVconsv12 alone did not protect against the SARS-CoV-2 infection, but when co-administered with 1/50th of the ChAdOx1 nCoV-19 spike vaccine protective dose, faster recovery and lower oral swab viral load were observed. Induction of CD8+ T cells may decrease COVID-19 severity and extend the T-cell response coverage of variants to match the known (and as yet unknown) members of the β-coronavirus family.
Collapse
Affiliation(s)
- Edmund G.-T. Wee
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK; (E.G.-T.W.); (A.C.)
| | - Sarah Kempster
- Science and Research—Diagnostics, Medicines and Healthcare products Regulatory Agency, Potters Bar EN6 3QG, UK; (S.K.); (D.F.); (J.H.); (C.H.); (N.A.)
| | - Deborah Ferguson
- Science and Research—Diagnostics, Medicines and Healthcare products Regulatory Agency, Potters Bar EN6 3QG, UK; (S.K.); (D.F.); (J.H.); (C.H.); (N.A.)
| | - Joanna Hall
- Science and Research—Diagnostics, Medicines and Healthcare products Regulatory Agency, Potters Bar EN6 3QG, UK; (S.K.); (D.F.); (J.H.); (C.H.); (N.A.)
| | - Claire Ham
- Science and Research—Diagnostics, Medicines and Healthcare products Regulatory Agency, Potters Bar EN6 3QG, UK; (S.K.); (D.F.); (J.H.); (C.H.); (N.A.)
| | - Susan Morris
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK (S.C.G.)
| | - Alison Crook
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK; (E.G.-T.W.); (A.C.)
| | - Sarah C. Gilbert
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK (S.C.G.)
| | - Bette Korber
- New Mexico Consortium, Los Alamos, NM 87544, USA;
| | - Neil Almond
- Science and Research—Diagnostics, Medicines and Healthcare products Regulatory Agency, Potters Bar EN6 3QG, UK; (S.K.); (D.F.); (J.H.); (C.H.); (N.A.)
| | - Tomáš Hanke
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK; (E.G.-T.W.); (A.C.)
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto 860-0811, Japan
| |
Collapse
|
10
|
Benhamouda N, Besbes A, Bauer R, Mabrouk N, Gadouas G, Desaint C, Chevrier L, Lefebvre M, Radenne A, Roelens M, Parfait B, Weiskopf D, Sette A, Gruel N, Courbebaisse M, Appay V, Paul S, Gorochov G, Ropers J, Lebbah S, Lelievre JD, Johannes L, Ulmer J, Lebeaux D, Friedlander G, De Lamballerie X, Ravel P, Kieny MP, Batteux F, Durier C, Launay O, Tartour E. Cytokine profile of anti-spike CD4 +T cells predicts humoral and CD8 +T cell responses after anti-SARS-CoV-2 mRNA vaccination. iScience 2024; 27:110441. [PMID: 39104410 PMCID: PMC11298648 DOI: 10.1016/j.isci.2024.110441] [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: 04/08/2024] [Revised: 05/31/2024] [Accepted: 07/01/2024] [Indexed: 08/07/2024] Open
Abstract
Coordinating immune responses - humoral and cellular - is vital for protection against severe Covid-19. Our study evaluates a multicytokine CD4+T cell signature's predictive for post-vaccinal serological and CD8+T cell responses. A cytokine signature composed of four cytokines (IL-2, TNF-α, IP10, IL-9) excluding IFN-γ, and generated through machine learning, effectively predicted the CD8+T cell response following mRNA-1273 or BNT162b2 vaccine administration. Its applicability extends to murine vaccination models, encompassing diverse immunization routes (such as intranasal) and vaccine platforms (including adjuvanted proteins). Notably, we found correlation between CD4+T lymphocyte-produced IL-21 and the humoral response. Consequently, we propose a test that offers a rapid overview of integrated immune responses. This approach holds particular relevance for scenarios involving immunocompromised patients because they often have low cell counts (lymphopenia) or pandemics. This study also underscores the pivotal role of CD4+T cells during a vaccine response and highlights their value in vaccine immunomonitoring.
Collapse
Affiliation(s)
- Nadine Benhamouda
- Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
- Université Paris Cité, INSERM U970, PARCC, Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
| | - Anissa Besbes
- Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
- Université Paris Cité, INSERM U970, PARCC, Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
| | | | - Nesrine Mabrouk
- Université Paris Cité, INSERM U970, PARCC, Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
| | - Gauthier Gadouas
- Bioinformatics and Cancer System Biology Team, IRCM-INSERM U1194, Institut de Recherche en Cancerologie de Montpellier, Montpellier, France
| | - Corinne Desaint
- INSERM SC10-US019, Villejuif, France
- Université Paris Cité, INSERM, CIC 1417, F-CRIN, Innovative Clinical Research Network in Vaccinology (I-REIVAC), APHP, CIC Cochin Pasteur, Hôpital Cochin, Paris, France
| | - Lucie Chevrier
- Université Paris Cité, INSERM U1016 Insitut Cochin, Hôpital Cochin, APHP, Centre Service d’immunologie Biologique, Paris, France
| | - Maeva Lefebvre
- Service de maladies infectieuses et tropicales, Centre de prévention des maladies infectieuses et transmissibles CHU de Nantes, Nantes, France
| | - Anne Radenne
- Unité de Recherche Clinique des Hôpitaux Universitaires Pitié Salpêtrière-Hôpitaux Universitaires Pitié Salpêtrière - Charles Foix, APHP, Paris, France
| | - Marie Roelens
- Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
- Université Paris Cité, INSERM U970, PARCC, Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
| | - Béatrice Parfait
- Centre de ressources Biologiques, Hôpital Cochin, APHP, Paris, France
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
- Department of Medicine, School of Medicine in Health Sciences, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Nadège Gruel
- INSERM U830, Équipe Labellisée Ligue Nationale Contre le Cancer, Diversity and Plasticity of Childhood Tumors Lab, Centre de Recherche, Institut Curie, Université PSL, Paris, France
- Department of Translational Research, Centre de Recherche, Institut Curie, Université PSL, Paris, France
| | - Marie Courbebaisse
- Faculté de Médecine, Université Paris Cité, Paris, France
- Explorations fonctionnelles rénales, Physiologie, Hôpital Européen Georges-Pompidou, APHP, Paris, France
| | - Victor Appay
- Université de Bordeaux, CNRS UMR 5164, INSERM ERL 1303, ImmunoConcEpT, 33000 Bordeaux, France
- International Research Center of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Stephane Paul
- Centre International de Recherche en Infectiologie, Team GIMAP, Université Jean Monnet, Université Claude Bernard Lyon, INSERM, CIC 1408 INSERM Vaccinology, Immunology Department, iBiothera Reference Center, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Guy Gorochov
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses, APHP, Hôpital Pitié-Salpêtrière, Paris, France
| | - Jacques Ropers
- Unité de Recherche Clinique des Hôpitaux Universitaires Pitié Salpêtrière –Hôpitaux Universitaires Pitié Salpêtrière- Charles Foix, APHP, Paris, France
| | - Said Lebbah
- Unité de Recherche Clinique des Hôpitaux Universitaires Pitié Salpêtrière –Hôpitaux Universitaires Pitié Salpêtrière- Charles Foix, APHP, Paris, France
| | - Jean-Daniel Lelievre
- Vaccine Research Institute, Créteil, France
- INSERM U955, Université Paris-Est Créteil, Créteil, France
- Groupe Henri-Mondor Albert-Chenevier, APHP, Créteil, France
| | - Ludger Johannes
- Cellular and Chemical Biology Unit, U1143 INSERM, UMR3666 CNRS, Institut Curie, Centre de Recherche, Université PSL, Paris, France
| | - Jonathan Ulmer
- Cellular and Chemical Biology Unit, U1143 INSERM, UMR3666 CNRS, Institut Curie, Centre de Recherche, Université PSL, Paris, France
| | - David Lebeaux
- Université Paris Cité, Service de maladies infectieuses Hôpital Saint Louis/Lariboisère APHP, INSERM, Paris, France
| | - Gerard Friedlander
- Department of « Croissance et Signalisation », Institut Necker Enfants Malades, INSERM U1151, CNRS UMR 8253, Université de Paris Cité, Paris, France
| | - Xavier De Lamballerie
- Unité des Virus Émergents, UVE: Aix-Marseille Université, IRD 190, INSERM 1207 Marseille, France
| | - Patrice Ravel
- Bioinformatics and Cancer System Biology Team, IRCM-INSERM U1194, Institut de Recherche en Cancerologie de Montpellier, Montpellier, France
| | - Marie Paule Kieny
- Institut National de la Santé et de la Recherche Médicale, INSERM, Paris, France
| | - Fréderic Batteux
- Université Paris Cité, INSERM U1016 Insitut Cochin, Hôpital Cochin, APHP, Centre Service d’immunologie Biologique, Paris, France
| | | | - Odile Launay
- Université Paris Cité, INSERM, CIC 1417, F-CRIN, Innovative Clinical Research Network in Vaccinology (I-REIVAC), APHP, CIC Cochin Pasteur, Hôpital Cochin, Paris, France
| | - Eric Tartour
- Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
- Université Paris Cité, INSERM U970, PARCC, Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
| |
Collapse
|
11
|
Binayke A, Zaheer A, Vishwakarma S, Sharma P, Dandotiya J, Raghavan S, Gosain M, Singh S, Chattopadhyay S, Kaushal J, Madan U, Kshetrapal P, Batra G, Wadhwa N, Pandey AK, Bhatnagar S, Garg PK, Awasthi A. Understanding the landscape of the SARS-CoV-2-specific T cells post-omicron surge. J Med Virol 2024; 96:e29877. [PMID: 39169721 DOI: 10.1002/jmv.29877] [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: 05/30/2024] [Revised: 07/24/2024] [Accepted: 08/13/2024] [Indexed: 08/23/2024]
Abstract
Emerging evidence shows increased humoral response post-omicron surge, but research on T cell responses is limited. This study investigated the durability, magnitude, and breadth of SARS-CoV-2-spike-specific T cell responses in 216 two-dose vaccinated individuals pre- and post-omicron surge. Post-surge samples showed enhanced T cell responses, indicating widespread asymptomatic exposure to omicron. Further analysis of 105 individuals with multiple exposures to SARS-CoV-2 through boosters or infections showed that post-omicron, two-dose vaccinated individuals had T cell responses comparable to those of COVID-19 convalescents or boosted individuals. Additionally, we report cross-reactive T cell responses against omicron sub-variants, including BA2.86, remained strong, with preserved frequencies of spike-specific stem-cell-like memory T cells. In silico prediction indicates that mutated epitopes of JN.1 and KP.2 retain over 95.6% of their HLA binding capability. Overall, our data suggests that T cell responses are sustained, enhanced, and cross-reactive against emerging SARS-CoV-2 variants following symptomatic or asymptomatic omicron infection.
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
| | - Priyanka Sharma
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Jyotsna Dandotiya
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, Faridabad, India
| | | | - Mudita Gosain
- Translational Health Science and Technology Institute, Faridabad, India
| | - Savita Singh
- Translational Health Science and Technology Institute, Faridabad, India
| | | | - Jyotsana Kaushal
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Upasna Madan
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, Faridabad, India
| | | | - Gaurav Batra
- Translational Health Science and Technology Institute, Faridabad, India
| | - Nitya Wadhwa
- Translational Health Science and Technology Institute, Faridabad, India
| | | | | | - Pramod Kumar Garg
- Translational Health Science and Technology Institute, Faridabad, India
- All India Institute of Medical Science, 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
|
12
|
Hodgson D, Liu Y, Carolan L, Mahanty S, Subbarao K, Sullivan SG, Fox A, Kucharski A. Memory B cell proliferation drives differences in neutralising responses between ChAdOx1 and BNT162b2 SARS-CoV-2 vaccines. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.11.24310221. [PMID: 39040163 PMCID: PMC11261961 DOI: 10.1101/2024.07.11.24310221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Vaccination against COVID-19 has been pivotal in reducing the global burden of the disease. However, Phase III trial results and observational studies underscore differences in efficacy across vaccine technologies and dosing regimens. Notably, mRNA vaccines have exhibited superior effectiveness compared to Adenovirus (AdV) vaccines, especially with extended dosing intervals. Using in-host mechanistic modelling, this study elucidates these variations and unravels the biological mechanisms shaping the immune responses at the cellular level. We used data on the change in memory B cells, plasmablasts, and antibody titres after the second dose of a COVID-19 vaccine for Australian healthcare workers. Alongside this dataset, we constructed a kinetic model of humoral immunity which jointly captured the dynamics of multiple immune markers, and integrated hierarchical effects into this kinetics model, including age, dosing schedule, and vaccine type. Our analysis estimated that mRNA vaccines induced 2.1 times higher memory B cell proliferation than AdV vaccines after adjusting for age, interval between doses and priming dose. Additionally, extending the duration between the second vaccine dose and priming dose beyond 28 days boosted neutralising antibody production per plasmablast concentration by 30%. We also found that antibody responses after the second dose were more persistent when mRNA vaccines were used over AdV vaccines and for longer dosing regimens. Reconstructing in-host kinetics in response to vaccination could help optimise vaccine dosing regimens, improve vaccine efficacy in different population groups, and inform the design of future vaccines for enhanced protection against emerging pathogens.
Collapse
Affiliation(s)
- David Hodgson
- Centre of Mathematical Modelling of Infectious Diseases, London School and Hygiene and Tropical Medicine, London, UK
| | - Yi Liu
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Louise Carolan
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Siddhartha Mahanty
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Kanta Subbarao
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Sheena G. Sullivan
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- School of Clinical Sciences, Monash University, Melbourne, Australia
| | - Annette Fox
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Adam Kucharski
- Centre of Mathematical Modelling of Infectious Diseases, London School and Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
13
|
Aguilar-Bretones M, den Hartog Y, van Dijk LLA, Malahe SRK, Dieterich M, Mora HT, Mueller YM, Koopmans MPG, Reinders MEJ, Baan CC, van Nierop GP, de Vries RD. SARS-CoV-2-specific immune responses converge in kidney disease patients and controls with hybrid immunity. NPJ Vaccines 2024; 9:93. [PMID: 38806532 PMCID: PMC11133345 DOI: 10.1038/s41541-024-00886-0] [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: 10/31/2023] [Accepted: 05/07/2024] [Indexed: 05/30/2024] Open
Abstract
Healthy individuals with hybrid immunity, due to a SARS-CoV-2 infection prior to first vaccination, have stronger immune responses compared to those who were exclusively vaccinated. However, little is known about the characteristics of antibody, B- and T-cell responses in kidney disease patients with hybrid immunity. Here, we explored differences between kidney disease patients and controls with hybrid immunity after asymptomatic or mild coronavirus disease-2019 (COVID-19). We studied the kinetics, magnitude, breadth and phenotype of SARS-CoV-2-specific immune responses against primary mRNA-1273 vaccination in patients with chronic kidney disease or on dialysis, kidney transplant recipients, and controls with hybrid immunity. Although vaccination alone is less immunogenic in kidney disease patients, mRNA-1273 induced a robust immune response in patients with prior SARS-CoV-2 infection. In contrast, kidney disease patients with hybrid immunity develop SARS-CoV-2 antibody, B- and T-cell responses that are equally strong or stronger than controls. Phenotypic analysis showed that Spike (S)-specific B-cells varied between groups in lymph node-homing and memory phenotypes, yet S-specific T-cell responses were phenotypically consistent across groups. The heterogeneity amongst immune responses in hybrid immune kidney patients warrants further studies in larger cohorts to unravel markers of long-term protection that can be used for the design of targeted vaccine regimens.
Collapse
Affiliation(s)
| | - Yvette den Hartog
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center Transplant Institute, Rotterdam, The Netherlands
| | - Laura L A van Dijk
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - S Reshwan K Malahe
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center Transplant Institute, Rotterdam, The Netherlands
| | - Marjolein Dieterich
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center Transplant Institute, Rotterdam, The Netherlands
| | - Héctor Tejeda Mora
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center Transplant Institute, Rotterdam, The Netherlands
| | - Yvonne M Mueller
- Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marion P G Koopmans
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marlies E J Reinders
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center Transplant Institute, Rotterdam, The Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center Transplant Institute, Rotterdam, The Netherlands
| | | | - Rory D de Vries
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
14
|
Brummelman J, Suárez-Hernández S, de Rond L, Bogaard-van Maurik M, Molenaar P, van Wijlen E, Oomen D, Beckers L, Rots NY, van Beek J, Nicolaie MA, van Els CACM, Boer MC, Kaaijk P, Buisman AM, de Wit J. Distinct T cell responsiveness to different COVID-19 vaccines and cross-reactivity to SARS-CoV-2 variants with age and CMV status. Front Immunol 2024; 15:1392477. [PMID: 38774878 PMCID: PMC11106399 DOI: 10.3389/fimmu.2024.1392477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/09/2024] [Indexed: 05/24/2024] Open
Abstract
Introduction Accumulating evidence indicates the importance of T cell immunity in vaccination-induced protection against severe COVID-19 disease, especially against SARS-CoV-2 Variants-of-Concern (VOCs) that more readily escape from recognition by neutralizing antibodies. However, there is limited knowledge on the T cell responses across different age groups and the impact of CMV status after primary and booster vaccination with different vaccine combinations. Moreover, it remains unclear whether age has an effect on the ability of T cells to cross-react against VOCs. Methods Therefore, we interrogated the Spike-specific T cell responses in healthy adults of the Dutch population across different ages, whom received different vaccine types for the primary series and/or booster vaccination, using IFNɣ ELISpot. Cells were stimulated with overlapping peptide pools of the ancestral Spike protein and different VOCs. Results Robust Spike-specific T cell responses were detected in the vast majority of participants upon the primary vaccination series, regardless of the vaccine type (i.e. BNT162b2, mRNA-1273, ChAdOx1 nCoV-19, or Ad26.COV2.S). Clearly, in the 70+ age group, responses were overall lower and showed more variation compared to younger age groups. Only in CMV-seropositive older adults (>70y) there was a significant inverse relation of age with T cell responses. Although T cell responses increased in all age groups after booster vaccination, Spike-specific T cell frequencies remained lower in the 70+ age group. Regardless of age or CMV status, primary mRNA-1273 vaccination followed by BNT162b2 booster vaccination showed limited booster effect compared to the BNT162b2/BNT162b2 or BNT162b2/mRNA-1273 primary-booster regimen. A modest reduction in cross-reactivity to the Alpha, Delta and Omicron BA.1, but not the Beta or Gamma variant, was observed after primary vaccination. Discussion Together, this study shows that age, CMV status, but also the primary-booster vaccination regimen influence the height of the vaccination-induced Spike-specific T cell response, but did not impact the VOC cross-reactivity.
Collapse
Affiliation(s)
- Jolanda Brummelman
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Sara Suárez-Hernández
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Lia de Rond
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Marjan Bogaard-van Maurik
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Petra Molenaar
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Emma van Wijlen
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Debbie Oomen
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Lisa Beckers
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Nynke Y. Rots
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Josine van Beek
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Mioara A. Nicolaie
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Cécile A. C. M. van Els
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
- Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Mardi C. Boer
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Patricia Kaaijk
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Anne-Marie Buisman
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Jelle de Wit
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| |
Collapse
|
15
|
Zhang J, Chang C, Liang Z, Hu T, Yin Z, Liang Y, Zhang T, Ding Y, Li X, Gai X, Yang X, Li X, Dong X, Ren J, Rao Y, Wang J, Yang J, Xue L, Sun Y. Elevated CD4 + T Cell Senescence Associates with Impaired Immune Responsiveness in Severe COVID-19. Aging Dis 2024; 16:AD.2024.0214-2. [PMID: 38377029 PMCID: PMC11745426 DOI: 10.14336/ad.2024.0214-2] [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: 10/11/2023] [Accepted: 02/14/2024] [Indexed: 02/22/2024] Open
Abstract
Aging is a critical risk factor for unfavorable clinical outcomes among COVID-19 patients and may impact vaccine efficacy. However, whether the senescence of T cells is associated with severe COVID-19 outcome in elderly individuals is unclear. Using flow cytometry, we analyzed the frequency of senescent T cells (Tsens) in peripheral blood from 100 hospitalized elderly COVID-19 patients and compared differences between those with mild/moderate and severe/critical illness. We also assessed correlations between the percentage of Tsens and the quantity and quality of spike-specific antibodies by ELISA, neutralizing antibody test kit, and ELISPOT assay respectively, the cytokine production profile of COVID-19 reactive T cells, and plasma soluble factors by cytometric bead array (CBA). Our study found a significantly elevated level of CD4+ Tsens in patients with severe/critical disease compared to those with mild/moderate illness. Patients with a higher level of CD4+ Tsens (>19.78%) showed a decreased survival rate compared to those with a lower level (≤19.78%). This is more pronounced among patients with breakthrough infections. The percentage of CD4+ Tsens was negatively correlated with spike-specific antibody titers, neutralization ability, and COVID-19 reactive IL-2+CD4+ T cells. In addition, spike-specific antibody levels were positively correlated with IL-2 producing T cells and plasma IL-2 amount. Mechanistically, with defective CD40L, T cells from patients with CD4+ Tsens >19.78% were unable to support B cell proliferation and differentiation. Our data demonstrate that the percentage of CD4+ Tsens in peripheral blood may serve as a reliable biomarker for the prognosis of severe COVID-19 patients, especially in breakthrough infections. Therefore, restoring the immune response of CD4+ Tsens may be key to preventing severe illness and improving vaccine efficacy in older adults.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China.
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
- Biobank, Peking University Third Hospital, Beijing, China.
| | - Chun Chang
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China.
| | - Zhaoyuan Liang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
| | - Tingting Hu
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China.
| | - Zhongnan Yin
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
- Biobank, Peking University Third Hospital, Beijing, China.
| | - Ying Liang
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China.
| | - Ting Zhang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
- Biobank, Peking University Third Hospital, Beijing, China.
| | - Yanling Ding
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China.
| | - Xianlong Li
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
| | - Xiaoyan Gai
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China.
| | - Xiaoxue Yang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
- Biobank, Peking University Third Hospital, Beijing, China.
| | - Xin Li
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China.
| | - Xixuan Dong
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
- Biobank, Peking University Third Hospital, Beijing, China.
| | - Jiaqi Ren
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China.
| | - Yafei Rao
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China.
| | - Jun Wang
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China.
| | - Jianling Yang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
| | - Lixiang Xue
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
- Biobank, Peking University Third Hospital, Beijing, China.
| | - Yongchang Sun
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
16
|
Nesamari R, Omondi MA, Baguma R, Höft MA, Ngomti A, Nkayi AA, Besethi AS, Magugu SFJ, Mosala P, Walters A, Clark GM, Mennen M, Skelem S, Adriaanse M, Grifoni A, Sette A, Keeton RS, Ntusi NAB, Riou C, Burgers WA. Post-pandemic memory T cell response to SARS-CoV-2 is durable, broadly targeted, and cross-reactive to the hypermutated BA.2.86 variant. Cell Host Microbe 2024; 32:162-169.e3. [PMID: 38211583 PMCID: PMC10901529 DOI: 10.1016/j.chom.2023.12.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/28/2023] [Accepted: 12/11/2023] [Indexed: 01/13/2024]
Abstract
Ongoing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) evolution has given rise to recombinant Omicron lineages that dominate globally (XBB.1), as well as the emergence of hypermutated variants (BA.2.86). In this context, durable and cross-reactive T cell immune memory is critical for continued protection against severe COVID-19. We examined T cell responses to SARS-CoV-2 approximately 1.5 years since Omicron first emerged. We describe sustained CD4+ and CD8+ spike-specific T cell memory responses in healthcare workers in South Africa (n = 39) who were vaccinated and experienced at least one SARS-CoV-2 infection. Spike-specific T cells are highly cross-reactive with all Omicron variants tested, including BA.2.86. Abundant nucleocapsid and membrane-specific T cells are detectable in most participants. The bulk of SARS-CoV-2-specific T cell responses have an early-differentiated phenotype, explaining their persistent nature. Overall, hybrid immunity leads to the accumulation of spike and non-spike T cells evident 3.5 years after the start of the pandemic, with preserved recognition of highly mutated SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Rofhiwa Nesamari
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Millicent A Omondi
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Richard Baguma
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Maxine A Höft
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Amkele Ngomti
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Anathi A Nkayi
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Asiphe S Besethi
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Siyabulela F J Magugu
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Paballo Mosala
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Avril Walters
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Gesina M Clark
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Mathilda Mennen
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa; Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; South African Medical Research Council Extramural Unit on Intersection of Non-communicable Disease and Infectious Diseases, University of Cape Town, Cape Town, South Africa
| | - Sango Skelem
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa; Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; South African Medical Research Council Extramural Unit on Intersection of Non-communicable Disease and Infectious Diseases, University of Cape Town, Cape Town, South Africa
| | - Marguerite Adriaanse
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa; Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; South African Medical Research Council Extramural Unit on Intersection of Non-communicable Disease and Infectious Diseases, University of Cape Town, Cape Town, South Africa
| | - Alba Grifoni
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Roanne S Keeton
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Ntobeko A B Ntusi
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa; Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; South African Medical Research Council Extramural Unit on Intersection of Non-communicable Disease and Infectious Diseases, University of Cape Town, Cape Town, South Africa; Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa
| | - Catherine Riou
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa; Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa.
| | - Wendy A Burgers
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa; Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
17
|
Höft MA, Burgers WA, Riou C. The immune response to SARS-CoV-2 in people with HIV. Cell Mol Immunol 2024; 21:184-196. [PMID: 37821620 PMCID: PMC10806256 DOI: 10.1038/s41423-023-01087-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 09/12/2023] [Indexed: 10/13/2023] Open
Abstract
This review examines the intersection of the HIV and SARS-CoV-2 pandemics. People with HIV (PWH) are a heterogeneous group that differ in their degree of immune suppression, immune reconstitution, and viral control. While COVID-19 in those with well-controlled HIV infection poses no greater risk than that for HIV-uninfected individuals, people with advanced HIV disease are more vulnerable to poor COVID-19 outcomes. COVID-19 vaccines are effective and well tolerated in the majority of PWH, though reduced vaccine efficacy, breakthrough infections and faster waning of vaccine effectiveness have been demonstrated in PWH. This is likely a result of suboptimal humoral and cellular immune responses after vaccination. People with advanced HIV may also experience prolonged infection that may give rise to new epidemiologically significant variants, but initiation or resumption of antiretroviral therapy (ART) can effectively clear persistent infection. COVID-19 vaccine guidelines reflect these increased risks and recommend prioritization for vaccination and additional booster doses for PWH who are moderately to severely immunocompromised. We recommend continued research and monitoring of PWH with SARS-CoV-2 infection, especially in areas with a high HIV burden.
Collapse
Affiliation(s)
- Maxine A Höft
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Wendy A Burgers
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa.
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa.
| | - Catherine Riou
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa.
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
18
|
Vecchio E, Rotundo S, Veneziano C, Abatino A, Aversa I, Gallo R, Giordano C, Serapide F, Fusco P, Viglietto G, Cuda G, Costanzo F, Russo A, Trecarichi EM, Torti C, Palmieri C. The spike-specific TCRβ repertoire shows distinct features in unvaccinated or vaccinated patients with SARS-CoV-2 infection. J Transl Med 2024; 22:33. [PMID: 38185632 PMCID: PMC10771664 DOI: 10.1186/s12967-024-04852-1] [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: 10/22/2023] [Accepted: 01/02/2024] [Indexed: 01/09/2024] Open
Abstract
BACKGROUND The evolving variants of SARS-CoV-2 may escape immunity from prior infections or vaccinations. It's vital to understand how immunity adapts to these changes. Both infection and mRNA vaccination induce T cells that target the Spike protein. These T cells can recognize multiple variants, such as Delta and Omicron, even if neutralizing antibodies are weakened. However, the degree of recognition can vary among people, affecting vaccine efficacy. Previous studies demonstrated the capability of T-cell receptor (TCR) repertoire analysis to identify conserved and immunodominant peptides with cross-reactive potential among variant of concerns. However, there is a need to extend the analysis of the TCR repertoire to different clinical scenarios. The aim of this study was to examine the Spike-specific TCR repertoire profiles in natural infections and those with combined natural and vaccine immunity. METHODS A T-cell enrichment approach and bioinformatic tools were used to investigate the Spike-specific TCRβ repertoire in peripheral blood mononuclear cells of previously vaccinated (n = 8) or unvaccinated (n = 6) COVID-19 patients. RESULTS Diversity and clonality of the TCRβ repertoire showed no significant differences between vaccinated and unvaccinated groups. When comparing the TCRβ data to public databases, 692 unique TCRβ sequences linked to S epitopes were found in the vaccinated group and 670 in the unvaccinated group. TCRβ clonotypes related to spike regions S135-177, S264-276, S319-350, and S448-472 appear notably more prevalent in the vaccinated group. In contrast, the S673-699 epitope, believed to have super antigenic properties, is observed more frequently in the unvaccinated group. In-silico analyses suggest that mutations in epitopes, relative to the main SARS-CoV-2 variants of concern, don't hinder their cross-reactive recognition by associated TCRβ clonotypes. CONCLUSIONS Our findings reveal distinct TCRβ signatures in vaccinated and unvaccinated individuals with COVID-19. These differences might be associated with disease severity and could influence clinical outcomes. TRIAL REGISTRATION FESR/FSE 2014-2020 DDRC n. 585, Action 10.5.12, noCOVID19@UMG.
Collapse
Affiliation(s)
- Eleonora Vecchio
- Department of Experimental and Clinical Medicine, University "Magna Graecia", Viale Europa, 88100, Catanzaro, Italy
- Interdepartmental Centre of Services, University "Magna Graecia", 88100, Catanzaro, Italy
| | - Salvatore Rotundo
- Department of Medical and Surgical Sciences, Chair of Infectious and Tropical Diseases, University "Magna Graecia", 88100, Catanzaro, Italy
| | - Claudia Veneziano
- Interdepartmental Centre of Services, University "Magna Graecia", 88100, Catanzaro, Italy
| | - Antonio Abatino
- Department of Experimental and Clinical Medicine, University "Magna Graecia", Viale Europa, 88100, Catanzaro, Italy
| | - Ilenia Aversa
- Department of Experimental and Clinical Medicine, University "Magna Graecia", Viale Europa, 88100, Catanzaro, Italy
| | - Raffaella Gallo
- Department of Experimental and Clinical Medicine, University "Magna Graecia", Viale Europa, 88100, Catanzaro, Italy
| | - Caterina Giordano
- Department of Experimental and Clinical Medicine, University "Magna Graecia", Viale Europa, 88100, Catanzaro, Italy
| | - Francesca Serapide
- Department of Medical and Surgical Sciences, Chair of Infectious and Tropical Diseases, University "Magna Graecia", 88100, Catanzaro, Italy
| | - Paolo Fusco
- Department of Medical and Surgical Sciences, Chair of Infectious and Tropical Diseases, University "Magna Graecia", 88100, Catanzaro, Italy
| | - Giuseppe Viglietto
- Department of Experimental and Clinical Medicine, University "Magna Graecia", Viale Europa, 88100, Catanzaro, Italy
| | - Giovanni Cuda
- Department of Experimental and Clinical Medicine, University "Magna Graecia", Viale Europa, 88100, Catanzaro, Italy
| | - Francesco Costanzo
- Department of Experimental and Clinical Medicine, University "Magna Graecia", Viale Europa, 88100, Catanzaro, Italy
- Interdepartmental Centre of Services, University "Magna Graecia", 88100, Catanzaro, Italy
| | - Alessandro Russo
- Department of Medical and Surgical Sciences, Chair of Infectious and Tropical Diseases, University "Magna Graecia", 88100, Catanzaro, Italy
| | - Enrico Maria Trecarichi
- Department of Medical and Surgical Sciences, Chair of Infectious and Tropical Diseases, University "Magna Graecia", 88100, Catanzaro, Italy
| | - Carlo Torti
- Department of Medical and Surgical Sciences, Chair of Infectious and Tropical Diseases, University "Magna Graecia", 88100, Catanzaro, Italy
| | - Camillo Palmieri
- Department of Experimental and Clinical Medicine, University "Magna Graecia", Viale Europa, 88100, Catanzaro, Italy.
| |
Collapse
|
19
|
Penetra SLS, Santos HFP, Resende PC, Bastos LS, da Silva MFB, Pina-Costa A, Lopes RS, Saboia-Vahia L, de Oliveira ACA, Pereira EC, Filho FM, Wakimoto MD, Calvet GA, Fuller TL, Whitworth J, Smith C, Nielsen-Saines K, Carvalho MS, Espíndola OM, Guaraldo L, Siqueira MM, Brasil P. SARS-CoV-2 Reinfection Cases in a Household-Based Prospective Cohort in Rio de Janeiro. J Infect Dis 2023; 228:1680-1689. [PMID: 37571849 PMCID: PMC11032242 DOI: 10.1093/infdis/jiad336] [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: 03/23/2023] [Revised: 08/02/2023] [Accepted: 08/09/2023] [Indexed: 08/13/2023] Open
Abstract
This was a household-based prospective cohort study conducted in Rio de Janeiro, in which people with laboratory-confirmed coronavirus disease 2019 (COVID-19) and their household contacts were followed from April 2020 through June 2022. Ninety-eight reinfections were identified, with 71 (72.5%) confirmed by genomic analyses and lineage definition in both infections. During the pre-Omicron period, 1 dose of any COVID-19 vaccine was associated with a reduced risk of reinfection, but during the Omicron period not even booster vaccines had this effect. Most reinfections were asymptomatic or milder in comparison with primary infections, a justification for continuing active surveillance to detect infections in vaccinated individuals. Our findings demonstrated that vaccination may not prevent infection or reinfection with severe acute respiratory syndrome coronavirus 2 (SARS CoV-2). Therefore we highlight the need to continuously update the antigenic target of SARS CoV-2 vaccines and administer booster doses to the population regularly, a strategy well established in the development of vaccines for influenza immunization programs.
Collapse
Affiliation(s)
- Stephanie L S Penetra
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Heloisa F P Santos
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paola Cristina Resende
- Laboratory of Respiratory Viruses and Measles National Influenza Centre, Americas Regional Reference Lab for Measles and Rubella, Reference Laboratory for COVID-19 World Health Organization, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Soares Bastos
- Scientific Computing Program, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Michele F B da Silva
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Anielle Pina-Costa
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renata Serrano Lopes
- Laboratory of Respiratory Viruses and Measles National Influenza Centre, Americas Regional Reference Lab for Measles and Rubella, Reference Laboratory for COVID-19 World Health Organization, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Saboia-Vahia
- Laboratory of Respiratory Viruses and Measles National Influenza Centre, Americas Regional Reference Lab for Measles and Rubella, Reference Laboratory for COVID-19 World Health Organization, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Any Caroline Alves de Oliveira
- Laboratory of Respiratory Viruses and Measles National Influenza Centre, Americas Regional Reference Lab for Measles and Rubella, Reference Laboratory for COVID-19 World Health Organization, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elisa Cavalcante Pereira
- Laboratory of Respiratory Viruses and Measles National Influenza Centre, Americas Regional Reference Lab for Measles and Rubella, Reference Laboratory for COVID-19 World Health Organization, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernando Medeiros Filho
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mayumi D Wakimoto
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Guilherme A Calvet
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Trevon L Fuller
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
- University of California, Los Angeles, Los Angeles, California, USA
| | - Jimmy Whitworth
- International Public Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Christopher Smith
- International Public Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
| | | | - Marilia Sá Carvalho
- Scientific Computing Program, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Otávio M Espíndola
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lusiele Guaraldo
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marilda M Siqueira
- Laboratory of Respiratory Viruses and Measles National Influenza Centre, Americas Regional Reference Lab for Measles and Rubella, Reference Laboratory for COVID-19 World Health Organization, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia Brasil
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
20
|
da Silva Antunes R, Weiskopf D, Sidney J, Rubiro P, Peters B, Arlehamn CSL, Grifoni A, Sette A. The MegaPool Approach to Characterize Adaptive CD4+ and CD8+ T Cell Responses. Curr Protoc 2023; 3:e934. [PMID: 37966108 PMCID: PMC10662678 DOI: 10.1002/cpz1.934] [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] [Indexed: 11/16/2023]
Abstract
Epitopes recognized by T cells are a collection of short peptide fragments derived from specific antigens or proteins. Immunological research to study T cell responses is hindered by the extreme degree of heterogeneity of epitope targets, which are usually derived from multiple antigens; within a given antigen, hundreds of different T cell epitopes can be recognized, differing from one individual to the next because T cell epitope recognition is restricted by the epitopes' ability to bind to MHC molecules, which are extremely polymorphic in different individuals. Testing large pools encompassing hundreds of peptides is technically challenging because of logistical considerations regarding solvent-induced toxicity. To address this issue, we developed the MegaPool (MP) approach based on sequential lyophilization of large numbers of peptides that can be used in a variety of assays to measure T cell responses, including ELISPOT, intracellular cytokine staining, and activation-induced marker assays, and that has been validated in the study of infectious diseases, allergies, and autoimmunity. Here, we describe the procedures for generating and testing MPs, starting with peptide synthesis and lyophilization, as well as a step-by-step guide and recommendations for their handling and experimental usage. Overall, the MP approach is a powerful strategy for studying T cell responses and understanding the immune system's role in health and disease. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Generation of peptide pools ("MegaPools") Basic Protocol 2: MegaPool testing and quantitation of antigen-specific T cell responses.
Collapse
Affiliation(s)
- Ricardo da Silva Antunes
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
| | - John Sidney
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
| | - Paul Rubiro
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
| | - Bjoern Peters
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| | | | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| |
Collapse
|
21
|
Silva-Moraes V, Souquette A, Sautto GA, Paciello I, Antonelli G, Andreano E, Rappuoli R, Teixeira-Carvalho A, Ross TM. Prior SARS-CoV-2 Infection Enhances Initial mRNA Vaccine Response with a Lower Impact on Long-Term Immunity. Immunohorizons 2023; 7:635-651. [PMID: 37819998 PMCID: PMC10615651 DOI: 10.4049/immunohorizons.2300041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/15/2023] [Indexed: 10/13/2023] Open
Abstract
Spike-encoding mRNA vaccines in early 2021 effectively reduced SARS-CoV-2-associated morbidity and mortality. New booster regimens were introduced due to successive waves of distinct viral variants. Therefore, people now have a diverse immune memory resulting from multiple SARS-CoV-2 Ag exposures, from infection to following vaccination. This level of community-wide immunity can induce immunological protection from SARS-CoV-2; however, questions about the trajectory of the adaptive immune responses and long-term immunity with respect to priming and repeated Ag exposure remain poorly explored. In this study, we examined the trajectory of adaptive immune responses following three doses of monovalent Pfizer BNT162b2 mRNA vaccination in immunologically naive and SARS-CoV-2 preimmune individuals without the occurrence of breakthrough infection. The IgG, B cell, and T cell Spike-specific responses were assessed in human blood samples collected at six time points between a moment before vaccination and up to 6 mo after the third immunization. Overall, the impact of repeated Spike exposures had a lower improvement on T cell frequency and longevity compared with IgG responses. Natural infection shaped the responses following the initial vaccination by significantly increasing neutralizing Abs and specific CD4+ T cell subsets (circulating T follicular helper, effector memory, and Th1-producing cells), but it had a small benefit at long-term immunity. At the end of the three-dose vaccination regimen, both SARS-CoV-2-naive and preimmune individuals had similar immune memory quality and quantity. This study provides insights into the durability of mRNA vaccine-induced immunological memory and the effects of preimmunity on long-term responses.
Collapse
Affiliation(s)
- Vanessa Silva-Moraes
- Center for Vaccines and Immunology, University of Georgia, Athens, GA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL
| | - Aisha Souquette
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Giuseppe A. Sautto
- Center for Vaccines and Immunology, University of Georgia, Athens, GA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL
| | - Ida Paciello
- Monoclonal Antibody Discovery Lab, Foundation Toscana Life Sciences, Siena, Italy
| | - Giada Antonelli
- Monoclonal Antibody Discovery Lab, Foundation Toscana Life Sciences, Siena, Italy
| | - Emanuele Andreano
- Monoclonal Antibody Discovery Lab, Foundation Toscana Life Sciences, Siena, Italy
| | | | | | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL
- Department of Infectious Diseases, University of Georgia, Athens, GA
- Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
22
|
Mak WA, Visser W, van der Vliet M, Markus HY, Koeleman JGM, Ong DSY. Ancestral SARS-CoV-2 and Omicron BA.5-specific neutralizing antibody and T-cell responses after Omicron bivalent booster vaccination in previously infected and infection-naive individuals. J Med Virol 2023; 95:e28989. [PMID: 37565645 DOI: 10.1002/jmv.28989] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/28/2023] [Accepted: 07/16/2023] [Indexed: 08/12/2023]
Abstract
Coronavirus disease-2019 (COVID-19) bivalent ancestral/Omicron messenger RNA (mRNA) booster vaccinations became available to boost and expand the immunity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron infections. In a prospective cohort study including 59 healthcare workers, we assessed SARS-CoV-2 ancestral and Omicron BA.5-specific neutralizing antibody and T-cell responses in previously infected and infection-naive individuals. Also, we assessed the effect of an ancestral/Omicron BA.1 bivalent mRNA booster vaccination on these immune responses. 10 months after previous monovalent mRNA vaccinations, ancestral SARS-CoV-2 S1-specific T-cell and anti-RBD IgG responses remained detectable in most individuals and a previous SARS-CoV-2 infection was associated with increased T-cell responses. T-cell responses, anti-RBD IgG, and Omicron BA.5 neutralization activity increased after receiving an ancestral/Omicron BA.1 bivalent booster mRNA vaccination. An Omicron BA.5 infection in addition to bivalent vaccination, led to a higher ratio of Omicron BA.5 to ancestral strain neutralization activity compared to no bivalent vaccination and no recent SARS-CoV-2 infection. In conclusion, SARS-CoV-2 T-cell and antibody responses persist for up to 10 months after a monovalent booster mRNA vaccination. An ancestral/Omicron BA.1 bivalent booster mRNA vaccination increases these immune responses and also induces Omicron BA.5 cross-neutralization antibody activity. Finally, our data indicate that hybrid immunity is associated with improved preservation of T-cell immunity.
Collapse
Affiliation(s)
- Willem A Mak
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Wendy Visser
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Marijke van der Vliet
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Hilde Y Markus
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Johannes G M Koeleman
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - David S Y Ong
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
23
|
Martel F, Cuervo-Rojas J, Ángel J, Ariza B, González JM, Ramírez-Santana C, Acosta-Ampudia Y, Murcia-Soriano L, Montoya N, Cardozo-Romero CC, Valderrama-Beltrán SL, Cepeda M, Castellanos JC, Gómez-Restrepo C, Perdomo-Celis F, Gazquez A, Dickson A, Brien JD, Mateus J, Grifoni A, Sette A, Weiskopf D, Franco MA. Cross-reactive humoral and CD4 + T cell responses to Mu and Gamma SARS-CoV-2 variants in a Colombian population. Front Immunol 2023; 14:1241038. [PMID: 37575243 PMCID: PMC10413264 DOI: 10.3389/fimmu.2023.1241038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 07/07/2023] [Indexed: 08/15/2023] Open
Abstract
The SARS CoV-2 antibody and CD4+ T cell responses induced by natural infection and/or vaccination decline over time and cross-recognize other viral variants at different levels. However, there are few studies evaluating the levels and durability of the SARS CoV-2-specific antibody and CD4+ T cell response against the Mu, Gamma, and Delta variants. Here, we examined, in two ambispective cohorts of naturally-infected and/or vaccinated individuals, the titers of anti-RBD antibodies and the frequency of SARS-CoV-2-specific CD4+ T cells up to 6 months after the last antigen exposure. In naturally-infected individuals, the SARS-CoV-2 antibody response declined 6 months post-symptoms onset. However, the kinetic observed depended on the severity of the disease, since individuals who developed severe COVID-19 maintained the binding antibody titers. Also, there was detectable binding antibody cross-recognition for the Gamma, Mu, and Delta variants, but antibodies poorly neutralized Mu. COVID-19 vaccines induced an increase in antibody titers 15-30 days after receiving the second dose, but these levels decreased at 6 months. However, as expected, a third dose of the vaccine caused a rise in antibody titers. The dynamics of the antibody response upon vaccination depended on the previous SARS-CoV-2 exposure. Lower levels of vaccine-induced antibodies were associated with the development of breakthrough infections. Vaccination resulted in central memory spike-specific CD4+ T cell responses that cross-recognized peptides from the Gamma and Mu variants, and their duration also depended on previous SARS-CoV-2 exposure. In addition, we found cross-reactive CD4+ T cell responses in unexposed and unvaccinated individuals. These results have important implications for vaccine design for new SARS-CoV-2 variants of interest and concern.
Collapse
Affiliation(s)
- Fabiola Martel
- Institute of Human Genetics, School of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Juliana Cuervo-Rojas
- Department of Clinical Epidemiology and Biostatistics, School of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Juana Ángel
- Institute of Human Genetics, School of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Beatriz Ariza
- Clinical Laboratory Science Research Group, Clinical Laboratory, Hospital Universitario San Ignacio, Bogotá, Colombia
| | - John Mario González
- Group of Basic Medical Sciences, School of Medicine, Universidad de Los Andes, Bogotá, Colombia
| | - Carolina Ramírez-Santana
- Center for Autoimmune Diseases Research
(CREA), School of Medicine and Health Sciences, Universidad del Rosario,, Bogotá, Colombia
| | - Yeny Acosta-Ampudia
- Center for Autoimmune Diseases Research
(CREA), School of Medicine and Health Sciences, Universidad del Rosario,, Bogotá, Colombia
| | | | - Norma Montoya
- Head Clinical Laboratory Unit, Clínica del Occidente, Bogotá, Colombia
| | | | - Sandra Liliana Valderrama-Beltrán
- Division of Infectious Diseases, Department of Internal Medicine. School of Medicine, Pontificia Universidad Javeriana, Hospital Universitario San Ignacio Infectious Diseases Research Group, Bogotá, Colombia
| | - Magda Cepeda
- Department of Clinical Epidemiology and Biostatistics, School of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | - Carlos Gómez-Restrepo
- Department of Clinical Epidemiology and Biostatistics, School of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Federico Perdomo-Celis
- Institute of Human Genetics, School of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Andreu Gazquez
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Alexandria Dickson
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - James D. Brien
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - José Mateus
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA, United States
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Manuel A. Franco
- Institute of Human Genetics, School of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
24
|
de Moor WRJ, Williamson AL, Schäfer G, Douglass N, Gers S, Sutherland AD, Blumenthal MJ, Margolin E, Shaw ML, Preiser W, Chapman R. LSDV-Vectored SARS-CoV-2 S and N Vaccine Protects against Severe Clinical Disease in Hamsters. Viruses 2023; 15:1409. [PMID: 37515096 PMCID: PMC10383203 DOI: 10.3390/v15071409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/17/2023] [Accepted: 06/18/2023] [Indexed: 07/30/2023] Open
Abstract
The SARS-CoV-2 pandemic demonstrated the need for potent and broad-spectrum vaccines. This study reports the development and testing of a lumpy skin disease virus (LSDV)-vectored vaccine against SARS-CoV-2, utilizing stabilized spike and conserved nucleocapsid proteins as antigens to develop robust immunogenicity. Construction of the vaccine (LSDV-SARS2-S,N) was confirmed by polymerase chain reaction (PCR) amplification and sequencing. In vitro characterization confirmed that cells infected with LSDV-SARS2-S,N expressed SARS-CoV-2 spike and nucleocapsid protein. In BALB/c mice, the vaccine elicited high magnitude IFN-γ ELISpot responses (spike: 2808 SFU/106 splenocytes) and neutralizing antibodies (ID50 = 6552). Testing in hamsters, which emulate human COVID-19 disease progression, showed the development of high titers of neutralizing antibodies against the Wuhan and Delta SARS-CoV-2 variants (Wuhan ID50 = 2905; Delta ID50 = 4648). Additionally, hamsters vaccinated with LSDV-SARS2-S,N displayed significantly less weight loss, lung damage, and reduced viral RNA copies following SARS-CoV-2 infection with the Delta variant as compared to controls, demonstrating protection against disease. These data demonstrate that LSDV-vectored vaccines display promise as an effective SARS-CoV-2 vaccine and as a potential vaccine platform for communicable diseases in humans and animals. Further efficacy testing and immune response analysis, particularly in non-human primates, are warranted.
Collapse
Affiliation(s)
- Warren R J de Moor
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Anna-Lise Williamson
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Georgia Schäfer
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
- International Centre for Genetic Engineering and Biotechnology, Observatory, Cape Town 7925, South Africa
- Wellcome Trust Centre for Infectious Disease Research in Africa, University of Cape Town, Cape Town 7925, South Africa
| | - Nicola Douglass
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | | | - Andrew D Sutherland
- Division of Medical Virology, Faculty of Medicine and Health Sciences, Stellenbosch University Tygerberg Campus, Cape Town 7505, South Africa
| | - Melissa J Blumenthal
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
- International Centre for Genetic Engineering and Biotechnology, Observatory, Cape Town 7925, South Africa
| | - Emmanuel Margolin
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
- Wellcome Trust Centre for Infectious Disease Research in Africa, University of Cape Town, Cape Town 7925, South Africa
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Cape Town 7701, South Africa
| | - Megan L Shaw
- Division of Medical Virology, Faculty of Medicine and Health Sciences, Stellenbosch University Tygerberg Campus, Cape Town 7505, South Africa
- Department of Medical Biosciences, Faculty of Natural Sciences, University of the Western Cape, Bellville, Cape Town 7535, South Africa
| | - Wolfgang Preiser
- Division of Medical Virology, Faculty of Medicine and Health Sciences, Stellenbosch University Tygerberg Campus, Cape Town 7505, South Africa
| | - Rosamund Chapman
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| |
Collapse
|
25
|
Liu Y, Sánchez-Ovando S, Carolan L, Dowson L, Khvorov A, Hadiprodjo J, Tseng YY, Delahunty C, Khatami A, Macnish M, Dougherty S, Hagenauer M, Riley KE, Jadhav A, Harvey J, Kaiser M, Mathew S, Hodgson D, Leung V, Subbarao K, Cheng AC, Macartney K, Koirala A, Marshall H, Clark J, Blyth CC, Wark P, Kucharski AJ, Sullivan SG, Fox A. Comparative B cell and antibody responses induced by adenoviral vectored and mRNA vaccines against COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.02.23290871. [PMID: 37333329 PMCID: PMC10275006 DOI: 10.1101/2023.06.02.23290871] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Both vector and mRNA vaccines were an important part of the response to the COVID-19 pandemic and may be required in future outbreaks and pandemics. However, adenoviral vectored (AdV) vaccines may be less immunogenic than mRNA vaccines against SARS-CoV-2. We assessed anti-spike and anti-vector immunity among infection-naïve Health Care Workers (HCW) following two doses of AdV (AZD1222) versus mRNA (BNT162b2) vaccine. 183 AdV and 274 mRNA vaccinees enrolled between April and October 2021. Median ages were 42 and 39 years, respectively. Blood was collected at least once, 10-48 days after vaccine dose 2. Surrogate virus neutralization test (sVNT) and spike binding antibody titres were a median of 4.2 and 2.2 times lower, respectively, for AdV compared to mRNA vaccinees (p<0.001). Median percentages of memory B cells that recognized fluorescent-tagged spike and RBD were 2.9 and 8.3 times lower, respectively for AdV compared to mRNA vaccinees. Titres of IgG reactive with human Adenovirus type 5 hexon protein rose a median of 2.2-fold after AdV vaccination but were not correlated with anti-spike antibody titres. Together the results show that mRNA induced substantially more sVNT antibody than AdV vaccine due to greater B cell expansion and targeting of the RBD. Pre-existing AdV vector cross-reactive antibodies were boosted following AdV vaccination but had no detectable effect on immunogenicity.
Collapse
Affiliation(s)
- Yi Liu
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Stephany Sánchez-Ovando
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Louise Carolan
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Leslie Dowson
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Arseniy Khvorov
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Jessica Hadiprodjo
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Yeu Yang Tseng
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Catherine Delahunty
- Immune Health Program, Hunter Medical Research Institute and School of Medicine and Public Health, University of Newcastle, Newcastle, Australia
| | - Ameneh Khatami
- The Children’s Hospital at Westmead; Sydney Children’s Hospital Network; National Centre for Immunisation Research and Surveillance, Sydney, Australia
- The University of Sydney; and National Centre for Immunisation Research and Surveillance, Sydney, Australia
| | - Marion Macnish
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Australia
| | - Sonia Dougherty
- Queensland Children’s Hospital, Children’s Health Queensland Hospital and Health Service; and University of Queensland, Brisbane, Australia
| | | | - Kathryn E. Riley
- Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Adelaide, Australia
- Division of Paediatric Medicine, Women’s and Children’s Health Network, Adelaide, Australia
| | - Ajay Jadhav
- The Children’s Hospital at Westmead; Sydney Children’s Hospital Network; National Centre for Immunisation Research and Surveillance, Sydney, Australia
| | - Joanne Harvey
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Australia
| | - Marti Kaiser
- Alfred Health and Monash University, Melbourne, Australia
| | - Suja Mathew
- Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Adelaide, Australia
- Division of Paediatric Medicine, Women’s and Children’s Health Network, Adelaide, Australia
| | - David Hodgson
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Vivian Leung
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Kanta Subbarao
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Allen C. Cheng
- Alfred Health and Monash University, Melbourne, Australia
| | - Kristine Macartney
- The Children’s Hospital at Westmead; Sydney Children’s Hospital Network; National Centre for Immunisation Research and Surveillance, Sydney, Australia
- The University of Sydney; and National Centre for Immunisation Research and Surveillance, Sydney, Australia
| | - Archana Koirala
- The Children’s Hospital at Westmead; Sydney Children’s Hospital Network; National Centre for Immunisation Research and Surveillance, Sydney, Australia
- The University of Sydney; and National Centre for Immunisation Research and Surveillance, Sydney, Australia
| | - Helen Marshall
- Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Adelaide, Australia
- Division of Paediatric Medicine, Women’s and Children’s Health Network, Adelaide, Australia
| | - Julia Clark
- Queensland Children’s Hospital, Children’s Health Queensland Hospital and Health Service; and University of Queensland, Brisbane, Australia
| | - Christopher C. Blyth
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Australia
- School of Medicine, University of Western Australia; Perth Children’s Hospital; and Department of Microbiology, PathWest Laboratory Medicine, QEII medical centre, Perth, Australia
| | - Peter Wark
- Immune Health Program, Hunter Medical Research Institute and School of Medicine and Public Health, University of Newcastle, Newcastle, Australia
| | - Adam J. Kucharski
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Sheena G. Sullivan
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Epidemiology, University of California, Los Angeles, USA
| | - Annette Fox
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| |
Collapse
|
26
|
Moyo-Gwete T, Richardson SI, Keeton R, Hermanus T, Spencer H, Manamela NP, Ayres F, Makhado Z, Motlou T, Tincho MB, Benede N, Ngomti A, Baguma R, Chauke MV, Mennen M, Adriaanse M, Skelem S, Goga A, Garrett N, Bekker LG, Gray G, Ntusi NA, Riou C, Burgers WA, Moore PL. Homologous Ad26.COV2.S vaccination results in reduced boosting of humoral responses in hybrid immunity, but elicits antibodies of similar magnitude regardless of prior infection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.15.23287288. [PMID: 36993404 PMCID: PMC10055608 DOI: 10.1101/2023.03.15.23287288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
The impact of previous SARS-CoV-2 infection on the durability of Ad26.COV2.S vaccine-elicited responses, and the effect of homologous boosting has not been well explored. We followed a cohort of healthcare workers for 6 months after receiving the Ad26.COV2.S vaccine and a further one month after they received an Ad26.COV2.S booster dose. We assessed longitudinal spike-specific antibody and T cell responses in individuals who had never had SARS-CoV-2 infection, compared to those who were infected with either the D614G or Beta variants prior to vaccination. Antibody and T cell responses elicited by the primary dose were durable against several variants of concern over the 6 month follow-up period, regardless of infection history. However, at 6 months after first vaccination, antibody binding, neutralization and ADCC were as much as 33-fold higher in individuals with hybrid immunity compared to those with no prior infection. Antibody cross-reactivity profiles of the previously infected groups were similar at 6 months, unlike at earlier time points suggesting that the effect of immune imprinting diminishes by 6 months. Importantly, an Ad26.COV2.S booster dose increased the magnitude of the antibody response in individuals with no prior infection to similar levels as those with previous infection. The magnitude of spike T cell responses and proportion of T cell responders remained stable after homologous boosting, concomitant with a significant increase in long-lived early differentiated CD4 memory T cells. Thus, these data highlight that multiple antigen exposures, whether through infection and vaccination or vaccination alone, result in similar boosts after Ad26.COV2.S vaccination.
Collapse
Affiliation(s)
- Thandeka Moyo-Gwete
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Simone I. Richardson
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Roanne Keeton
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology; University of Cape Town; Observatory, South Africa
| | - Tandile Hermanus
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Holly Spencer
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Nelia P. Manamela
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Frances Ayres
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Zanele Makhado
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Thopisang Motlou
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Marius B. Tincho
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology; University of Cape Town; Observatory, South Africa
| | - Ntombi Benede
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology; University of Cape Town; Observatory, South Africa
| | - Amkele Ngomti
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology; University of Cape Town; Observatory, South Africa
| | - Richard Baguma
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology; University of Cape Town; Observatory, South Africa
| | - Masego V. Chauke
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology; University of Cape Town; Observatory, South Africa
| | - Mathilda Mennen
- Department of Medicine, University of Cape Town and Groote Schuur Hospital; Observatory, South Africa
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town; Observatory, South Africa
- South African Medical Research Council Extramural Unit on Intersection of Non-communicable Diseases and Infectious Diseases, University of Cape Town, Cape Town, South Africa
| | - Marguerite Adriaanse
- Department of Medicine, University of Cape Town and Groote Schuur Hospital; Observatory, South Africa
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town; Observatory, South Africa
- South African Medical Research Council Extramural Unit on Intersection of Non-communicable Diseases and Infectious Diseases, University of Cape Town, Cape Town, South Africa
| | - Sango Skelem
- Department of Medicine, University of Cape Town and Groote Schuur Hospital; Observatory, South Africa
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town; Observatory, South Africa
- South African Medical Research Council Extramural Unit on Intersection of Non-communicable Diseases and Infectious Diseases, University of Cape Town, Cape Town, South Africa
| | - Ameena Goga
- South African Medical Research Council, Cape Town, South Africa
| | - Nigel Garrett
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
- Discipline of Public Health Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Linda-Gail Bekker
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Desmond Tutu HIV Centre, Cape Town, South Africa
| | - Glenda Gray
- South African Medical Research Council, Cape Town, South Africa
| | - Ntobeko A.B. Ntusi
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Department of Medicine, University of Cape Town and Groote Schuur Hospital; Observatory, South Africa
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town; Observatory, South Africa
- South African Medical Research Council Extramural Unit on Intersection of Non-communicable Diseases and Infectious Diseases, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Observatory, South Africa
| | - Catherine Riou
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology; University of Cape Town; Observatory, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Observatory, South Africa
| | - Wendy A. Burgers
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology; University of Cape Town; Observatory, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Observatory, South Africa
| | - Penny L. Moore
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
| |
Collapse
|