1
|
Accrombessi M, Dangbemey P, Assongba L, Yadouleton A, Dangbenon E, Wakpo N, Akogbeto MC, Protopopoff N, Cook J, Hounkpatin B. Previous or current infection with SARS-CoV-2 virus and its impact on maternal and neonatal health outcomes in Benin: a sero-epidemiological study in pregnant women. Arch Public Health 2025; 83:143. [PMID: 40481557 PMCID: PMC12142973 DOI: 10.1186/s13690-025-01633-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/17/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND SARS-CoV-2 (COVID-19) has emerged as a significant global public health challenge, revealing critical vulnerabilities within health systems worldwide. While extensive data on COVID-19 is available from high-income countries, information remains scarce in lower-income regions, particularly regarding its impact on pregnant women. This study aims to evaluate the burden of COVID-19 among pregnant women and its effects on maternal and birth outcomes during the third wave in Benin. METHODS A cross-sectional, hospital-based survey was conducted from May 19 to September 19, 2022, at the Lagune Mother and Child Teaching Hospital. A standardized questionnaire was administered, and nasal swabs along with serological analysis were performed on 437 pregnant women. Multivariate logistic regression was used to assess risk factors and evaluate the impact of previous or current COVID-19 exposure on maternal and birth adverse outcomes. RESULTS SARS-CoV-2 was detected in less than 1% of pregnant women through PCR testing of nasal swab samples. Among the study population, 14.4% of women were vaccinated against COVID-19. A total of 81.1% of women tested positive for antibodies, suggesting prior exposure or infection to SARS-CoV-2 or vaccination. Notably, 78.6% of unvaccinated women had detectable antibodies, which serves as a more accurate proxy for infection prevalence. No significant association was found between prior COVID-19 exposure and adverse maternal and birth outcomes (aOR: 0.48, 95% CI 0.15-1.51). CONCLUSIONS Although PCR testing revealed a low incidence of active SARS-CoV-2 infection, the high prevalence of IgG antibodies among pregnant women suggests widespread prior exposure or infection. Vaccination was identified as a strong predictor of detectable IgG antibodies. Notably, despite the presence of antibodies, no significant association was found between prior COVID-19 exposure and adverse maternal or birth outcomes. These findings highlight the need for further research to explore the potential long-term effects of COVID-19 infection on pregnancy outcomes and to better understand the relationship between antibody presence and maternal and fetal health.
Collapse
Affiliation(s)
- Manfred Accrombessi
- Centre de Recherche Entomologique de Cotonou, Cotonou, Benin.
- Institut de Recherche Clinique du Benin (IRCB), Abomey-Calavi, Benin.
| | - Patrice Dangbemey
- Department of Obstetrics and Gynaecology, Lagoon Mother and Child University Hospital Center, Faculty of Health Science, University of Abomey Calavi, Cotonou, Benin
| | - Landry Assongba
- Centre de Recherche Entomologique de Cotonou, Cotonou, Benin
| | - Anges Yadouleton
- Laboratory of Viral Haemorrhagic Fevers and Arboviruses of Benin, Ministry of Health, Cotonou, Benin
- Ecole Normale Supérieure de Natitingou, Technology, Engineering and Mathematics, National University of Science, Abomey, Benin
| | | | - Nelly Wakpo
- Department of Obstetrics and Gynaecology, Lagoon Mother and Child University Hospital Center, Faculty of Health Science, University of Abomey Calavi, Cotonou, Benin
| | | | - Natacha Protopopoff
- Department of Epidemiology and Public Health, Swiss Tropical & Public Health Institute, Allschwill, Switzerland
- University of Basel, Basel, Switzerland
| | - Jackie Cook
- International Statistics and Epidemiology Group, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Benjamin Hounkpatin
- Department of Obstetrics and Gynaecology, Lagoon Mother and Child University Hospital Center, Faculty of Health Science, University of Abomey Calavi, Cotonou, Benin
| |
Collapse
|
2
|
Sarjomaa M, Berg KK, Jaioun K, Tveten Y, Kersten H, Reiso H, Eikeland R, Thilesen C, Nordbø SA, Aaberge IS, Pearce N, Fell AKM. SARS-CoV-2-specific humoral immunity in a Norwegian cohort between 2020 and 2023. BMC Med 2025; 23:332. [PMID: 40462062 PMCID: PMC12135409 DOI: 10.1186/s12916-025-04171-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 05/27/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND We have previously reported on natural humoral immunity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in a Norwegian cohort between 2020 and 2021. In this study, we evaluated long-term humoral (including vaccination-induced) immunity in the same cohort and assessed predictors of high antibody levels against spike protein, as well as the persistence of antibodies against the virus spike and nucleocapsid proteins. METHODS Vaccination data and antibody levels against the spike and nucleocapsid proteins were collected at 12 (only in infected participants) and 24 months (in both infected and uninfected participants) after the participants' first polymerase chain reaction (PCR) tests for the virus. Antibody levels against spike protein at 24 months were categorized as high or low based on the 50th percentile. Possible predictors of high antibody levels against spike protein were examined using univariate and multivariate logistic regression models. RESULTS Of 1119 original participants (400 PCR + and 719 PCR -), 574 responded to our questionnaires and were invited to antibody measurements (median age: 51 years; women: 59%). Vaccination data showed that 11% were fully immunized, and 85% were booster-immunized at 24 months. Antibody levels were evaluated in 72% (287/400) of the PCR + participants at 12 months and 58% (233/400) at 24 months. At 12 and 24 months, we observed that 97% (278/287) and 100% (233/233), respectively, still had antibodies against the spike protein, and 86% (248/287) and 95% (221/233), respectively, against the nucleocapsid protein. Antibody levels were also evaluated in 34% (247/719) of those in the PCR - group, which revealed that 99.5% and 69% had detectable antibodies against spike and nucleocapsid proteins, respectively, at 24 months. Irrespective of pre-vaccination SARS-CoV-2 infection status, the booster-immunized participants were 3.7 × more likely to have high antibody levels against spike protein vs the non-booster-immunized ones. Those aged > 60 years had the highest median antibody levels against the spike protein and were more likely to be booster-immunized. CONCLUSIONS Our findings highlight the benefits of booster vaccinations for humoral immune responses. Long-term antibody levels against the SARS-CoV-2 spike protein were higher in booster-immunized participants vs the non-booster-immunized, irrespective of pre-vaccination infection status. TRIAL REGISTRATION 146,469: The COVID-19 study in Telemark and Agder-COVITA. CLINICALTRIALS gov ID: NCT04514003.
Collapse
Affiliation(s)
- Marjut Sarjomaa
- Department of Infection Control, Telemark Hospital Trust, Ulefossvegen 55, 3710, Skien, Norway.
- Department of Community Medicine and Global Health, University of Oslo, Oslo, Norway.
| | | | - Keson Jaioun
- Department of Research, Telemark Hospital Trust, Skien, Norway
- Department of Occupational and Environmental Medicine, Telemark Hospital Trust, Skien, Norway
| | - Yngvar Tveten
- Department of Clinical Microbiology, Telemark Hospital Trust, Skien, Norway
| | - Hege Kersten
- Department of Research, Telemark Hospital Trust, Skien, Norway
- Department of Geriatric Medicine, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Harald Reiso
- The Norwegian Advisory Unit On Tick-Borne Diseases, Sørlandet Hospital Trust, Arendal, Norway
| | - Randi Eikeland
- The Norwegian Advisory Unit On Tick-Borne Diseases, Sørlandet Hospital Trust, Arendal, Norway
- Department of Health and Sport Science, Institute of Health and Nursing Science, University of Agder, Grimstad, Norway
| | | | - Svein Arne Nordbø
- Department of Medical Microbiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Neil Pearce
- London School of Hygiene and Tropical Medicine, London, UK
| | - Anne Kristin Moeller Fell
- Department of Community Medicine and Global Health, University of Oslo, Oslo, Norway
- Department of Occupational and Environmental Medicine, Telemark Hospital Trust, Skien, Norway
| |
Collapse
|
3
|
Paul MJ, Hudda MT, Pallett S, Groppelli E, Boariu E, Finardi NF, Wake R, Sofat N, Biddle K, Koushesh S, Dwyer-Hemmings L, Cook R, Ma JKC. Mucosal immune responses to SARS-CoV-2 infection and COVID-19 vaccination. Vaccine 2025; 56:127175. [PMID: 40311214 DOI: 10.1016/j.vaccine.2025.127175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/20/2025] [Accepted: 04/21/2025] [Indexed: 05/03/2025]
Abstract
SARS-CoV-2 continues to circulate in the community. We hypothesise that mucosal immunity is required to prevent continuing viral acquisition and transmission. OBJECTIVES To determine whether SARS-CoV-2 infection or vaccination elicits specific neutralising antibodies in saliva, and to assess the longevity of protection. METHODS Initially, 111 COVID-19 convalescent participants were recruited, 11-369 days after diagnosis. Saliva and blood samples were assayed for antibodies specific for Spike protein, Receptor Binding Domain and Nucleoprotein. In a second cohort, 123 participants were recruited. Saliva and serum antibodies to the same antigens were assayed before and after their first and second COVID-19 vaccinations, with 150 day follow up. RESULTS Natural infection induces and boosts IgA and IgG in oral fluid and serum; vaccination does not induce or boost specific saliva IgA; IgG can be found in saliva after vaccination, but only when serum IgG concentrations are high; IgA is important for SARS-CoV-2 neutralisation activity by oral fluid, but there can also be contributions from serum IgG and other factors. CONCLUSIONS New COVID-19 vaccines should target both systemic and mucosal immunity, to establish a first line of immune defence at the mucosal barrier. This would benefit vulnerable patient populations and may help to eradicate SARS-CoV-2 circulation.
Collapse
Affiliation(s)
- Mathew J Paul
- Institute for Infection and Immunity, City St. George's University of London, Cranmer Terrace, London SW17 0RE, UK.
| | - Mohammed T Hudda
- Department of Population Health, Dasman Institute, Jasim Mohamad Al Bahar St, Kuwait City, Kuwait.
| | - Scott Pallett
- Institute for Infection and Immunity, City St. George's University of London, Cranmer Terrace, London SW17 0RE, UK; Centre of Defence Pathology, Royal Centre of Defence Medicine, Queen Elizabeth Hospital Birmingham, Birmingham, B15 2WB, UK; St. George's University Hospitals NHS Foundation Trust, Blackshaw Road Tooting, London SW17 0QT, UK.
| | - Elisabetta Groppelli
- Institute for Infection and Immunity, City St. George's University of London, Cranmer Terrace, London SW17 0RE, UK.
| | - Eugenia Boariu
- Institute for Infection and Immunity, City St. George's University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Nicole Falci Finardi
- Institute for Infection and Immunity, City St. George's University of London, Cranmer Terrace, London SW17 0RE, UK.
| | - Rachel Wake
- Institute for Infection and Immunity, City St. George's University of London, Cranmer Terrace, London SW17 0RE, UK; St. George's University Hospitals NHS Foundation Trust, Blackshaw Road Tooting, London SW17 0QT, UK.
| | - Nidhi Sofat
- Institute for Infection and Immunity, City St. George's University of London, Cranmer Terrace, London SW17 0RE, UK; St. George's University Hospitals NHS Foundation Trust, Blackshaw Road Tooting, London SW17 0QT, UK.
| | - Kathryn Biddle
- Institute for Infection and Immunity, City St. George's University of London, Cranmer Terrace, London SW17 0RE, UK; St. George's University Hospitals NHS Foundation Trust, Blackshaw Road Tooting, London SW17 0QT, UK.
| | - Soraya Koushesh
- Institute for Infection and Immunity, City St. George's University of London, Cranmer Terrace, London SW17 0RE, UK.
| | - Louis Dwyer-Hemmings
- Institute for Infection and Immunity, City St. George's University of London, Cranmer Terrace, London SW17 0RE, UK; St. George's University Hospitals NHS Foundation Trust, Blackshaw Road Tooting, London SW17 0QT, UK
| | - Richard Cook
- Faculty of Dentistry, Oral & Craniofacial Sciences, Kings College London, Floor 22, Guy's Tower, Guy's Hospital, Great Maze Pond, London, SE1 1UL, UK.
| | - Julian K-C Ma
- Institute for Infection and Immunity, City St. George's University of London, Cranmer Terrace, London SW17 0RE, UK; St. George's University Hospitals NHS Foundation Trust, Blackshaw Road Tooting, London SW17 0QT, UK.
| |
Collapse
|
4
|
Hasan Z, Masood KI, Qaiser S, Kanji A, Mwenda F, Alenquer M, Iqbal J, Ferreira F, Wassan Y, Balouch S, Yameen M, Hussain S, Begum K, Feroz K, Muhammad S, Sadiqa A, Akhtar M, Habib A, Ahmed SMA, Mian AA, Hussain R, Amorim MJ, Bhutta ZA. Comparative study of humoral and cellular immunity against SARS-CoV-2 induced by different COVID-19 vaccine types: Insights into protection against wildtype, Delta and JN.1 omicron strains. Vaccine 2025; 59:127270. [PMID: 40408899 DOI: 10.1016/j.vaccine.2025.127270] [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: 02/19/2025] [Revised: 04/19/2025] [Accepted: 05/12/2025] [Indexed: 05/25/2025]
Abstract
We investigated the effectiveness of different COVID-19 vaccinations administered in Pakistan by studying the effect of inactivated virus, mRNA and vector formulations. This study in 916 participants was conducted between October 2021 and July 2022. Subjects receiving inactivated (A), mRNA (B), one-dose vector (C), and two-dose vector (D) vaccines were sampled at baseline, 6, 12, and 24 weeks. Serum IgG antibodies to wildtype Spike and its receptor binding domain (RBD) were measured. Pseudovirus particle-based neutralizing assays against wildtype, Delta, and JN.1 variants were performed. T cell IFN-γ responses to SARS-CoV-2 antigens were measured. Participants were aged 37.05 ± 14.44 years and comprised 48.6 % females. Baseline Spike seropositivity rose from 90 % to 96 % by 24 weeks; and 40 % to 90 % against RBD. Group B participants had the highest anti-RBD levels which peaked by 6 weeks. IgG RBD in group A and C increased up until 24 weeks. Anti-RBD levels were reduced in those over 50 years. At baseline neutralizing titers were present at 38.5 % against wildtype and in 34.2 % against Delta variants. Titers doubled in vaccine groups A-C by 12 weeks, with highest titers in B and lowest in group C participants. At baseline, neutralizing titers against the JN.1 variant were absent but low titers were evident in 10 % of participants after 12 weeks. T cell reactivity to SARS-CoV-2 increased from 31 % at baseline to 50 % in group A and 73 % in group B participants by 6 weeks after vaccination. Presence of immunity against wildtype and Delta variants in one-third of participants at baseline could be due to sub-clinical infections. Increase in humoral and cellular immunity was greater after mRNA as compared with inactivated vaccinations. As COVID-19 morbidity in the population remained low, our data supports effectiveness of multiple vaccine formulations in protecting against severe COVID-19 in this high transmission population.
Collapse
Affiliation(s)
- Zahra Hasan
- Department of Pathology and Laboratory Medicine(1), The Aga Khan University (AKU), Karachi, Pakistan.
| | - Kiran Iqbal Masood
- Department of Pathology and Laboratory Medicine(1), The Aga Khan University (AKU), Karachi, Pakistan
| | - Shama Qaiser
- Department of Pathology and Laboratory Medicine(1), The Aga Khan University (AKU), Karachi, Pakistan
| | - Akbar Kanji
- Department of Pathology and Laboratory Medicine(1), The Aga Khan University (AKU), Karachi, Pakistan
| | - Fridah Mwenda
- Department of Pathology and Laboratory Medicine(1), The Aga Khan University (AKU), Karachi, Pakistan
| | - Marta Alenquer
- Católica Biomedical Research Centre, Católica Medical School, Universidade Católica Portuguesa, Palma de Cima, 1649-023 Lisboa, Portugal
| | - Junaid Iqbal
- Center of Excellence in Women and Child Health, AKU, Karachi, Pakistan
| | - Filipe Ferreira
- Católica Biomedical Research Centre, Católica Medical School, Universidade Católica Portuguesa, Palma de Cima, 1649-023 Lisboa, Portugal
| | - Yaqub Wassan
- Center of Excellence in Women and Child Health, AKU, Karachi, Pakistan
| | - Sadaf Balouch
- Department of Pathology and Laboratory Medicine(1), The Aga Khan University (AKU), Karachi, Pakistan
| | - Maliha Yameen
- Department of Pathology and Laboratory Medicine(1), The Aga Khan University (AKU), Karachi, Pakistan
| | - Shahneel Hussain
- Center of Excellence in Women and Child Health, AKU, Karachi, Pakistan
| | - Kehkashan Begum
- Center of Excellence in Women and Child Health, AKU, Karachi, Pakistan
| | - Khalid Feroz
- Center of Excellence in Women and Child Health, AKU, Karachi, Pakistan
| | - Sajid Muhammad
- Center of Excellence in Women and Child Health, AKU, Karachi, Pakistan
| | - Ayesha Sadiqa
- Department of Pathology and Laboratory Medicine(1), The Aga Khan University (AKU), Karachi, Pakistan
| | - Mishgan Akhtar
- Department of Pathology and Laboratory Medicine(1), The Aga Khan University (AKU), Karachi, Pakistan
| | - Atif Habib
- Center of Excellence in Women and Child Health, AKU, Karachi, Pakistan
| | | | - Afsar Ali Mian
- Center for Regenerative Medicine, AKU, Karachi, Pakistan
| | - Rabia Hussain
- Department of Pathology and Laboratory Medicine(1), The Aga Khan University (AKU), Karachi, Pakistan
| | - Maria Joao Amorim
- Católica Biomedical Research Centre, Católica Medical School, Universidade Católica Portuguesa, Palma de Cima, 1649-023 Lisboa, Portugal
| | - Zulfiqar A Bhutta
- Center of Excellence in Women and Child Health, AKU, Karachi, Pakistan; Centre for Global Child Health, Hospital for Sick Children, Toronto, Canada
| |
Collapse
|
5
|
Liu C, Tsang TK, Sullivan SG, Cowling BJ, Yang B. Comparative duration of neutralizing responses and protections of COVID-19 vaccination and correlates of protection. Nat Commun 2025; 16:4748. [PMID: 40404724 PMCID: PMC12098666 DOI: 10.1038/s41467-025-60024-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 05/06/2025] [Indexed: 05/24/2025] Open
Abstract
The decline in neutralizing antibody (nAb) titers and vaccine efficacy /effectiveness (VE) for SARS-CoV-2 vaccines has been observed over time and when confronted with emerging variants, two factors that are hard to distinguish. Despite substantial drop in nAb titers against Omicron, VE remains high for severe cases and fatalities, raising questions about the utility of detected nAbs as a correlate of protection for COVID-19 vaccines for varying disease severity. Here, we conducted a systematic comparison of waning dynamics of nAb and VE over time and against variants with varying levels of disease severity. Using Bayesian linear regression models, we found that antigenically-shifted variants, like Omicron, could potentially lead to greater reductions in nAb titers and primary VE against mild infections than associated immunity waning observed over a 180-day period. By comparing model predicted nAb titers and VE on the same time scales, we found that VE against severe and fatal outcomes remained above 75% even when nAb titers reached the detectable limit of assays, despite strong correlations with nAb titers (spearman correlations ≥0.7) across variants over time. This finding suggested detectable nAb titers are not always sensitive enough to fully predict protection against severe disease and death from SARS-CoV-2.
Collapse
Affiliation(s)
- Chang Liu
- World Health Organization Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
| | - Tim K Tsang
- World Health Organization Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, Hong Kong, China
| | - Sheena G Sullivan
- School of Clinical Sciences, Monash University, Melbourne, Australia
- Department of Epidemiology, University of California, Los Angeles, USA
| | - Benjamin J Cowling
- World Health Organization Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, Hong Kong, China
| | - Bingyi Yang
- World Health Organization Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China.
| |
Collapse
|
6
|
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
|
7
|
Yao Z, Feng Z, Zhang H, Zhang B. ScRNA-Seq reveals T cell immunity in COVID-19 patients and implications for immunotherapy. Int Immunopharmacol 2025; 155:114663. [PMID: 40233451 DOI: 10.1016/j.intimp.2025.114663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 03/26/2025] [Accepted: 04/09/2025] [Indexed: 04/17/2025]
Abstract
SARS-CoV-2, the virus causing COVID-19, poses significant health threats due to its high transmissibility and potential for severe respiratory complications. T cells, central to adaptive immunity, also interact with innate immunity, playing a pivotal role in coordinating defenses and eliminating infected cells. Single-cell RNA sequencing (scRNA-seq) has provided more subtle heterogeneity, rare subpopulations, or new subpopulations that are at the district differentiation stage or with specific function. Thus, elucidating how T cell heterogeneity impacts COVID-19 disease severity remains a critical question requiring comprehensive analysis. This review revealed the heterogeneity of the host T cells, including conventional T cells (CD8+, CD4+ T cells) and unconventional T cells, including natural killer T (NKT) cells, mucosal-associated invariant T (MAIT) and gamma-delta T (γδT) cells in COVID-19 patients with different clinical manifestations. Severe COVID-19 had marked lymphopenia, excessive activation, elevated exhaustion and reduced functional diversity of T cells. Pathogenic contributions arise from dysregulated cytotoxic T cells, Treg cells and unconventional T cells collectively driving systemic hyperinflammation and tissue injury. Current therapeutic strategies targeting T cells-such as enhancing virus-specific T cell responses, reverting T-cell exhaustion and alleviating inflammation-exhibit inconsistent efficacy, underscoring the need for combinatorial approaches. This review highlights how scRNA-seq deciphers T cell heterogeneity and dysfunction in COVID-19. By targeting T cell exhaustion, inflammation, and subset-specific deficits, these insights pave the way for therapies and vaccines.
Collapse
Affiliation(s)
- Zhihong Yao
- Faculty of Clinical Medicine, Hanzhong Vocational and Technical College, Hanzhong 723002, China; Affiliated Hospital, Hanzhong Vocational and Technical College, Hanzhong 723012, China; Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Zhao Feng
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Hui Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, NHC Key Laboratory of AIDS Prevention and Treatment, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, China Medical University, Shenyang, China.
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
8
|
Ha MK, Postovskaya A, Kuznetsova M, Meysman P, Van Deuren V, Van Ierssel S, De Reu H, Schippers J, Peeters K, Besbassi H, Heyndrickx L, Willems B, Mariën J, Bartholomeus E, Vercauteren K, Beutels P, Van Damme P, Lion E, Vlieghe E, Laukens K, Coenen S, Naesens R, Ariën KK, Ogunjimi B. Celluloepidemiology-A paradigm for quantifying infectious disease dynamics on a population level. SCIENCE ADVANCES 2025; 11:eadt2926. [PMID: 40378227 DOI: 10.1126/sciadv.adt2926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 04/15/2025] [Indexed: 05/18/2025]
Abstract
To complement serology as a tool in public health interventions, we introduced the "celluloepidemiology" paradigm where we leveraged pathogen-specific T cell responses at a population level to advance our epidemiological understanding of infectious diseases, using SARS-CoV-2 as a model. Applying flow cytometry and machine learning on data from more than 500 individuals, we showed that the number of T cells with positive expression of functional markers not only could distinguish patients who recovered from COVID-19 from controls and pre-COVID donors but also identify previously unrecognized asymptomatic patients from mild, moderate, and severe recovered patients. The celluloepidemiology approach was uniquely capable to differentiate health care worker groups with different SARS-CoV-2 exposures from each other. T cell receptor (TCR) profiling strengthened our analysis by revealing that SARS-CoV-2-specific TCRs were more abundant in patients than in controls. We believe that adding data on T cell reactivity will complement serology and augment the value of infection morbidity modeling for populations.
Collapse
Affiliation(s)
- My K Ha
- Center for Health Economics Research and Modelling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
| | - Anna Postovskaya
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- ADReM Data Lab, Department of Mathematics and Computer Science, University of Antwerp, Antwerp, Belgium
- Biomedical Informatics Research Network Antwerp (biomina), University of Antwerp, Antwerp, Belgium
- Clinical Virology Unit, Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Maria Kuznetsova
- Center for Health Economics Research and Modelling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
| | - Pieter Meysman
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- ADReM Data Lab, Department of Mathematics and Computer Science, University of Antwerp, Antwerp, Belgium
- Biomedical Informatics Research Network Antwerp (biomina), University of Antwerp, Antwerp, Belgium
| | - Vincent Van Deuren
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- ADReM Data Lab, Department of Mathematics and Computer Science, University of Antwerp, Antwerp, Belgium
- Biomedical Informatics Research Network Antwerp (biomina), University of Antwerp, Antwerp, Belgium
| | - Sabrina Van Ierssel
- Department of General Internal Medicine, Infectious Disease and Tropical Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Hans De Reu
- Laboratory of Experimental Hematology (LEH), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Flow Cytometry and Cell Sorting Core Facility (FACSUA), University of Antwerp, Wilrijk, Belgium
| | - Jolien Schippers
- Center for Health Economics Research and Modelling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
| | - Karin Peeters
- Center for Health Economics Research and Modelling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
| | - Hajar Besbassi
- Center for Health Economics Research and Modelling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
| | - Leo Heyndrickx
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Betty Willems
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Joachim Mariën
- Department of Ecology and Evolutionary Biology, University of Antwerp, Antwerp, Belgium
- The Virus Ecology Group, Institute of Tropical Medicine, Antwerp, Belgium
| | - Esther Bartholomeus
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
| | - Koen Vercauteren
- Clinical Virology Unit, Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Philippe Beutels
- Center for Health Economics Research and Modelling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
| | - Pierre Van Damme
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- Centre for the Evaluation of Vaccination (CEV), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
| | - Eva Lion
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- Laboratory of Experimental Hematology (LEH), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Flow Cytometry and Cell Sorting Core Facility (FACSUA), University of Antwerp, Wilrijk, Belgium
| | - Erika Vlieghe
- Department of General Internal Medicine, Infectious Disease and Tropical Medicine, Antwerp University Hospital, Edegem, Belgium
- Global Health Institute, University of Antwerp, Wilrijk, Belgium
| | - Kris Laukens
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- ADReM Data Lab, Department of Mathematics and Computer Science, University of Antwerp, Antwerp, Belgium
- Biomedical Informatics Research Network Antwerp (biomina), University of Antwerp, Antwerp, Belgium
| | - Samuel Coenen
- Laboratory of Medical Microbiology (LMM), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Wilrijk, Belgium
- Center for General Practice, Department of Family Medicine and Population Health (FAMPOP), University of Antwerp, Wilrijk, Belgium
| | - Reinout Naesens
- Department of Clinical Biology, Antwerp Hospital Network, Antwerp, Belgium
| | - Kevin K Ariën
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Benson Ogunjimi
- Center for Health Economics Research and Modelling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- Department of Pediatrics, Antwerp University Hospital, Edegem, Belgium
| |
Collapse
|
9
|
Luo Q, Song Q, Li Y, Zong K, Liu T, He J, Mei G, Du H, Xia Z, Liu M, Song J, Gao C, Xia D, Xue G, Tian W, Qu Y, Kou Z, Dong Z, Han J. Reduced immune response to SARS-CoV-2 infection in the elderly after 6 months. Front Immunol 2025; 16:1596065. [PMID: 40416973 PMCID: PMC12098630 DOI: 10.3389/fimmu.2025.1596065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Accepted: 04/11/2025] [Indexed: 05/27/2025] Open
Abstract
Objectives To evaluate the immune persistence and cross-immune response of elderly individuals after Omicron BA.5 infections. Method The neutralizing antibodies against WT, BA.5, XBB.1 and EG.5 strains were analyzed. The T/B-cell subsets' responses were tested through intracellular cytokine staining and flow cytometry. Results The neutralizing antibodies titers against WT and BA.5 strain, remaining high level for at least 6 months, were higher than that of both XBB.1 and EG.5 variants. The neutralizing antibodies of WT, BA.5, XBB.1, and EG.5 strains in the elderly were slightly lower than those in middle-age. The memory B cells decreased rapidly in the elderly, and Tfh, Th17 cells of the elderly continued to increase for only 3 months, while Tfh and Th17 cells increased in the middle-aged for over 6 months. For the elderly, after peptide stimulation, unswitched/switched memory B cells decreased, while double negative B cells displayed higher proliferation. The proportions of both naïve and Temra cells in CD4+ and CD8+ T cells declined, whereas those of Tcm and Tem cells elevated. In the meantime, both CD69+ and CD38+ T cells decreased, but the frequencies of PD-1+ and CTLA-4+ of CD4+ and CD8+ T cells showed an increasing trend. The proportions of PD-1+ and CTLA-4+ cells also increased in older people with long COVID symptoms at 3m post-infection. Conclusions Omicron BA.5 infection induced lower neutralizing antibodies against XBB.1 and EG.5 variant. The decrease of memory B cells, CD69+ and CD38+T cells, as well as the increase of PD-1+, CTLA-4+ of CD4+/CD8+T cells and double negative B cells, indicate that sustained immune responses against BA.5 infection may wane more rapidly in elderly populations.
Collapse
Affiliation(s)
- Qin Luo
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Qinqin Song
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yan Li
- Shandong Center for Disease Control and Prevention, Shandong Provincial Key Laboratory of Intelligent Monitoring, Early Warning, and Prevention and Control of Infectious Diseases, Shandong Institute of Preventive Medicine, Jinan, China
| | - Kexin Zong
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ti Liu
- Shandong Center for Disease Control and Prevention, Shandong Provincial Key Laboratory of Intelligent Monitoring, Early Warning, and Prevention and Control of Infectious Diseases, Shandong Institute of Preventive Medicine, Jinan, China
| | - Junming He
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Guoyong Mei
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Haijun Du
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhiqiang Xia
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Mi Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Juan Song
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Chen Gao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Dong Xia
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Guangyu Xue
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Wenyan Tian
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yinli Qu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zengqiang Kou
- Shandong Center for Disease Control and Prevention, Shandong Provincial Key Laboratory of Intelligent Monitoring, Early Warning, and Prevention and Control of Infectious Diseases, Shandong Institute of Preventive Medicine, Jinan, China
| | - Zhongjun Dong
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical Immunology, Anhui Medical University, Hefei, China
- State Key Laboratory of Membrane Biology, School of Medicine and Institute for Immunology, Tsinghua University, Beijing, China
| | - Jun Han
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
10
|
Mayola Danés N, Brownlie D, Folkman R, Nordlander A, Blom K, Varnaite R, Niessl J, Karlsson Lindsjö O, Söderholm S, Akber M, Chen P, Buggert M, Bråve A, Klingström J, Nowak P, Marquardt N, Sondén K, Blennow O, Gredmark-Russ S. Dysregulated Adaptive Immune Responses to SARS-CoV-2 in Immunocompromised Individuals. Microorganisms 2025; 13:1077. [PMID: 40431250 PMCID: PMC12114339 DOI: 10.3390/microorganisms13051077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/27/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
The SARS-CoV-2 virus poses a significant risk to immunocompromised patients, who display weakened immunity and reduced seroconversion following infection and vaccination. In this study, we recruited 19 hospitalized patients with immune disorders (ImCo) and 4 immunocompetent controls (ICC) with COVID-19. We evaluated their serological, humoral, and cellular immune responses at <30 days and >90 days post-symptom onset. ICC patients showed robust B and T cell responses against SARS-CoV-2, indicated by detectable antibody levels, memory antibody-secreting cells (mASCs) towards the spike protein and spike-specific CD4+ and CD8+ T cells. ImCo patients showed impaired immune responses, with lower levels of B cell responses. Further subdivision of the ImCo patients demonstrates that solid organ transplant (SOT) patients generated B cell responses similar to ICC patients, whereas the other ImCo patients, including patients with hematological malignancies and anti-CD20 therapy, did not. Absolute T cell numbers and spike-specific CD4+ and CD8+ T cell responses were low in the ImCo patients at <30 days but increased at later time points. Our findings suggest that even when B cell responses were reduced, patients could present a T cell response, suggesting a more successful line of passive immunization for immunocompromised individuals focusing on boosting T cell responses.
Collapse
Affiliation(s)
- Núria Mayola Danés
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden
| | - Demi Brownlie
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden
| | - Rebecca Folkman
- Department of Infectious Diseases, Karolinska University Hospital, 141 86 Stockholm, Sweden
| | - Anna Nordlander
- Department of Infectious Diseases, Karolinska University Hospital, 141 86 Stockholm, Sweden
- Department of Laboratory Medicine, Karolinska Institutet, 141 52 Huddinge, Sweden
| | - Kim Blom
- Public Health Agency of Sweden, 171 65 Solna, Sweden
- Department of Clinical Microbiology, Umeå University, 901 87 Umeå, Sweden
| | - Renata Varnaite
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden
| | - Julia Niessl
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden
| | | | | | - Mira Akber
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden
| | - Puran Chen
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden
| | - Marcus Buggert
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden
| | - Andreas Bråve
- Public Health Agency of Sweden, 171 65 Solna, Sweden
| | - Jonas Klingström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden
- Public Health Agency of Sweden, 171 65 Solna, Sweden
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, 581 83 Linköping, Sweden
| | - Piotr Nowak
- Department of Infectious Diseases, Karolinska University Hospital, 141 86 Stockholm, Sweden
- Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden
| | - Nicole Marquardt
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden
| | - Klara Sondén
- Public Health Agency of Sweden, 171 65 Solna, Sweden
- Department of Medicine Solna, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Ola Blennow
- Department of Infectious Diseases, Karolinska University Hospital, 141 86 Stockholm, Sweden
- Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden
| | - Sara Gredmark-Russ
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, 141 86 Stockholm, Sweden
- Laboratory for Molecular Infection Medicine Sweden, Umeå University, 901 87 Umeå, Sweden
| |
Collapse
|
11
|
Vaňová V, Náhliková J, Ličková M, Sláviková M, Kajanová I, Lukáčiková Ľ, Sabo M, Rádiková Ž, Pastoreková S, Klempa B. Long-Term Dynamics of SARS-CoV-2 Variant-Specific Neutralizing Antibodies Following mRNA Vaccination and Infection. Viruses 2025; 17:675. [PMID: 40431687 PMCID: PMC12115524 DOI: 10.3390/v17050675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/29/2025] Open
Abstract
Understanding the long-term dynamics of SARS-CoV-2 neutralizing antibodies is critical for evaluating vaccine-induced protection and informing booster strategies. In this longitudinal study, we analyzed 114 serum samples from 19 individuals across six time points over a three-year period following mRNA vaccination (Comirnaty) and natural SARS-CoV-2 infection. Using pseudotype-based neutralization assays against nine SARS-CoV-2 variants, including major Omicron subvariants (BA.1-BA.5, BQ.1.1, XBB), and anti-S1 IgG ELISA, we observed that antibody levels peaked after the third vaccine dose and remained relatively stable two years later. Neutralization titers rose markedly after the second and third doses, with the highest neutralization observed at two years post-booster. Strong correlations were found between anti-S1 IgG levels and mean neutralization titers for pre-Omicron variants (r = 0.79-0.93; p < 0.05), but only moderate for Omicron subvariants (r ≈ 0.50-0.64). Notably, hybrid immunity (vaccination plus infection) resulted in higher neutralization titers at the final time point compared to vaccine-only participants. The lowest neutralization was observed against XBB, underscoring the immune evasiveness of emerging variants. These findings support the importance of booster vaccination and highlight the added durability of hybrid immunity in long-term protection.
Collapse
Affiliation(s)
- Veronika Vaňová
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (V.V.); (J.N.); (M.L.); (M.S.); (I.K.); (Ľ.L.); (S.P.)
| | - Jana Náhliková
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (V.V.); (J.N.); (M.L.); (M.S.); (I.K.); (Ľ.L.); (S.P.)
| | - Martina Ličková
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (V.V.); (J.N.); (M.L.); (M.S.); (I.K.); (Ľ.L.); (S.P.)
| | - Monika Sláviková
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (V.V.); (J.N.); (M.L.); (M.S.); (I.K.); (Ľ.L.); (S.P.)
| | - Ivana Kajanová
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (V.V.); (J.N.); (M.L.); (M.S.); (I.K.); (Ľ.L.); (S.P.)
| | - Ľubomíra Lukáčiková
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (V.V.); (J.N.); (M.L.); (M.S.); (I.K.); (Ľ.L.); (S.P.)
| | - Miroslav Sabo
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia;
| | - Žofia Rádiková
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia;
| | - Silvia Pastoreková
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (V.V.); (J.N.); (M.L.); (M.S.); (I.K.); (Ľ.L.); (S.P.)
| | - Boris Klempa
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (V.V.); (J.N.); (M.L.); (M.S.); (I.K.); (Ľ.L.); (S.P.)
| |
Collapse
|
12
|
Edeling MA, Earnest L, Carrera Montoya J, Yap AHY, Mumford J, Roberts J, Wong CY, Hans D, Grima J, Bisset N, Bodle J, Rockman S, Torresi J. Development of Methods to Produce SARS CoV-2 Virus-Like Particles at Scale. Biotechnol Bioeng 2025; 122:1118-1129. [PMID: 39936889 PMCID: PMC11975197 DOI: 10.1002/bit.28937] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 02/13/2025]
Abstract
The devastating global toll precipitated by the SARS CoV-2 outbreak and the profound impact of vaccines in stemming that outbreak has established the need for molecular platforms capable of rapidly delivering effective, safe and accessible medical interventions in pandemic preparedness. We describe a simple, efficient and adaptable process to produce SARS CoV-2 virus-like particles (VLPs) that can be readily scaled for manufacturing. A rapid but gentle method of tangential flow filtration using a 100 kDa semi-permeable membrane concentrates and buffer exchanges 0.5 L of SARS CoV-2 VLP containing supernatant into low salt and optimal pH for anion exchange chromatography. VLPs are washed, eluted under high salt, dialyzed into physiological buffer, sterile filtered and aliquoted for storage at -80°C. Purification is completed in less than 2 days. A simple quality control process includes Western blot for coincident detection of Spike, Membrane and Envelope protein as a proxy for intact VLP, ELISA to detect conformationally sensitive Spike using readily available anti-Spike and/or anti-RBD antibodies, and negative stain and immunogold electron microscopy to validate particulate, Spike crowned VLPs. This process to produce SARS CoV-2 VLPs for preclinical studies serves as a roadmap for preparation of more distantly related VLPs for pandemic preparedness.
Collapse
Affiliation(s)
- Melissa A. Edeling
- Department of Microbiology and ImmunologyThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| | - Linda Earnest
- Department of Microbiology and ImmunologyThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| | - Julio Carrera Montoya
- Department of Microbiology and ImmunologyThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| | - Ashley Huey Yiing Yap
- Department of Microbiology and ImmunologyThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| | - Jamie Mumford
- Victorian Infectious Diseases Reference laboratoryRoyal Melbourne Hospital at the Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| | - Jason Roberts
- Victorian Infectious Diseases Reference laboratoryRoyal Melbourne Hospital at the Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
- Department of Infectious DiseasesThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| | - Chinn Yi Wong
- Department of Microbiology and ImmunologyThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| | - Dhiraj Hans
- Research, Innovation & Commercialisation, Faculty of Medicine, Dentistry & Health SciencesThe University of MelbourneParkvilleVictoriaAustralia
| | - Joseph Grima
- Seqirus, Vaccine Innovation UnitParkvilleVictoriaAustralia
| | - Nicole Bisset
- Seqirus, Vaccine Innovation UnitParkvilleVictoriaAustralia
| | - Jesse Bodle
- Seqirus, Vaccine Innovation UnitParkvilleVictoriaAustralia
| | - Steven Rockman
- Seqirus, Vaccine Innovation UnitParkvilleVictoriaAustralia
| | - Joseph Torresi
- Department of Microbiology and ImmunologyThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| |
Collapse
|
13
|
Mcconney CS, Kenney D, Ennis CS, Smith-Mahoney EL, Ayuso MJ, Zhong J, Douam F, Sagar M, Snyder-Cappione JE. Individuals Infected with SARS-CoV-2 Prior to COVID-19 Vaccination Maintain Vaccine-Induced RBD-Specific Antibody Levels and Viral Neutralization Activity for One Year. Viruses 2025; 17:640. [PMID: 40431652 PMCID: PMC12115583 DOI: 10.3390/v17050640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/30/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
The effectiveness of multiple COVID-19 vaccinations in individuals with a history of SARS-CoV-2 infection remains unclear; specifically, elucidation of the durability of anti-viral antibody responses could provide important insights for epidemiological applications. We utilized the BU ELISA protocol to measure the circulating SARS-CoV-2 receptor-binding domain (RBD) and nucleocapsid (N) specific IgG and IgA antibody levels in a cohort of individuals infected with SARS-CoV-2 in the spring of 2020, with the sample collection spanning six months to two years post-symptom onset. Further, we interrogated the neutralization activity of these samples against the ancestral SARS-CoV-2 (WA-1) and Delta and Omicron (BA.1) variants. Consistent with previous studies, we found a more rapid waning of anti-N compared to anti-RBD antibodies in months prior to the first vaccinations. Vaccine-induced antibody responses in individuals previously infected with SARS-CoV-2 were elevated and sustained for more than one year post-vaccination. Similarly, neutralization activity against WA-1, Delta, and Omicron increased and remained higher than pre-vaccination levels for one year after the first COVID-19 vaccine dose. Collectively, these results indicate that infection followed by vaccination yields robust antibody responses against SARS-CoV-2 that endure for one year. These results suggest that an annual booster would stably boost anti-SARS-CoV-2 antibody responses, preventing infection and disease.
Collapse
Affiliation(s)
- Christina S. Mcconney
- Department of Virology, Immunology, and Microbiology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA; (C.S.M.); (D.K.); (M.J.A.); (J.Z.); (F.D.)
| | - Devin Kenney
- Department of Virology, Immunology, and Microbiology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA; (C.S.M.); (D.K.); (M.J.A.); (J.Z.); (F.D.)
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02118, USA
| | - Christina S. Ennis
- Cancer Center, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA;
| | - Erika L. Smith-Mahoney
- Department of Virology, Immunology, and Microbiology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA; (C.S.M.); (D.K.); (M.J.A.); (J.Z.); (F.D.)
| | - Maria Jose Ayuso
- Department of Virology, Immunology, and Microbiology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA; (C.S.M.); (D.K.); (M.J.A.); (J.Z.); (F.D.)
| | - Jiabao Zhong
- Department of Virology, Immunology, and Microbiology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA; (C.S.M.); (D.K.); (M.J.A.); (J.Z.); (F.D.)
| | - Florian Douam
- Department of Virology, Immunology, and Microbiology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA; (C.S.M.); (D.K.); (M.J.A.); (J.Z.); (F.D.)
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02118, USA
| | - Manish Sagar
- Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA;
| | - Jennifer E. Snyder-Cappione
- Department of Virology, Immunology, and Microbiology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA; (C.S.M.); (D.K.); (M.J.A.); (J.Z.); (F.D.)
| |
Collapse
|
14
|
Yılmaz S, Eken A, Sezer Z, Bağcı BŞ, Erdem S, Sarıkaya MD, Kaplan B, Inal A, Bayram A, Kalın Unuvar G, Zararsız G, Yerlitas Sİ, Cakir N, Pavel STI, Uygut MA, Yetiskin H, Kara A, Ozdarendeli A. Vaccination with inactivated SARS-CoV-2 vaccine TURKOVAC induces durable humoral and cellular immune responses up to 8 months. Front Med (Lausanne) 2025; 12:1524393. [PMID: 40357274 PMCID: PMC12066321 DOI: 10.3389/fmed.2025.1524393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/31/2025] [Indexed: 05/15/2025] Open
Abstract
Background The rapid spread of the SARS-CoV-2 virus has led to a global health crisis, necessitating swift responses in medical science, mainly through vaccination strategies. While short-term vaccine effectiveness is evident, immune protection's long-term effects and duration remain incompletely understood. Systematic monitoring of these responses is essential for optimizing vaccination strategies. Aims This study aimed to explore the durability of antigen-specific T and B cell responses and antibody levels up to 8 months post-immunization with the inactivated TURKOVAC vaccine in volunteers. Additionally, the impact of two versus three doses of vaccination on these parameters was analyzed. Methods Volunteers (n = 80) received two or three doses of TURKOVAC. Spike-specific B cells, CD4+ T cells, CD8+ T cells, and antibody levels were measured at multiple time points post-immunization. Results Spike-specific B cells remained elevated up to 8 months post-immunization. SARS-CoV-2-specific CD4+ and CD8+ T cells peaked at 4 months but declined thereafter. TURKOVAC resulted in durable antigen-specific humoral and cellular immune memory with distinct kinetics. Still, most assessments observed no significant differences between two and three doses, except for antigen specific-IL-2 and CD4+ LAMP1 responses. Conclusion TURKOVAC vaccination induces durable immune responses, with spike-specific B cells persisting up to 8 months and T cell responses peaking at 4 months before declining. These findings suggest that TURKOVAC contributes to long-term immune protection against SARS-CoV-2.
Collapse
Affiliation(s)
- Seçil Yılmaz
- Genome and Stem Cell Center, Erciyes University, Kayseri, Türkiye
| | - Ahmet Eken
- Genome and Stem Cell Center, Erciyes University, Kayseri, Türkiye
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Zafer Sezer
- Department of Medical Pharmacology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
- Good Clinical Practise Centre (IKUM), Erciyes University, Kayseri, Türkiye
| | - Burcu Şen Bağcı
- Vaccine Research, Development and Application Centre (ERAGEM), Erciyes University, Kayseri, Türkiye
| | - Serife Erdem
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | | | - Busra Kaplan
- Vaccine Research, Development and Application Centre (ERAGEM), Erciyes University, Kayseri, Türkiye
- Department of Medical Microbiology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Ahmet Inal
- Department of Medical Pharmacology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
- Good Clinical Practise Centre (IKUM), Erciyes University, Kayseri, Türkiye
| | - Adnan Bayram
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Gamze Kalın Unuvar
- Infectious Diseases Clinic, Department of Infectious Diseases, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Gokmen Zararsız
- Department of Biostatistics, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Serra İlayda Yerlitas
- Department of Biostatistics, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Nuri Cakir
- Department of Medical Microbiology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | | | - Muhammet Ali Uygut
- Vaccine Research, Development and Application Centre (ERAGEM), Erciyes University, Kayseri, Türkiye
| | - Hazel Yetiskin
- Vaccine Research, Development and Application Centre (ERAGEM), Erciyes University, Kayseri, Türkiye
| | - Ates Kara
- Pediatric Infectious Department, Faculty of Medicine, Hacettepe University Hospitals, Ankara, Türkiye
| | - Aykut Ozdarendeli
- Vaccine Research, Development and Application Centre (ERAGEM), Erciyes University, Kayseri, Türkiye
- Department of Medical Microbiology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| |
Collapse
|
15
|
Molinos-Albert LM, Rubio R, Martín-Pérez C, Pradenas E, Torres C, Jiménez A, Canyelles M, Vidal M, Barrios D, Marfil S, Aparicio E, Ramírez-Morros A, Trinité B, Vidal-Alaball J, Santamaria P, Serra P, Izquierdo L, Aguilar R, Ruiz-Comellas A, Blanco J, Dobaño C, Moncunill G. Long-lasting antibody B-cell responses to SARS-CoV-2 three years after the onset of the pandemic. Cell Rep 2025; 44:115498. [PMID: 40173043 DOI: 10.1016/j.celrep.2025.115498] [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: 09/09/2024] [Revised: 12/21/2024] [Accepted: 03/11/2025] [Indexed: 04/04/2025] Open
Abstract
Immune memory is essential for the effectiveness of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination. In the current context of the pandemic, with a diminished vaccine efficacy against emerging variants, it remains crucial to perform long-term studies to evaluate the durability and quality of immune responses. Here, we examined the antibody and memory B-cell responses in a cohort of 113 healthcare workers with distinct exposure histories over a 3-year period. Previously infected and naive participants developed comparable humoral responses by 17 months after receiving a full three-dose mRNA vaccination. In addition, both maintained a substantial SARS-CoV-2-reactive memory B-cell pool, associated with a lower incidence of breakthrough infections in naive participants. Of note, previously infected participants developed an expanded SARS-CoV-2-reactive CD27-CD21- atypical B-cell population that remained stable throughout the follow-up period. Thus, previous SARS-CoV-2 infection differentially imprints the memory B-cell compartment without compromising the development of long-lasting humoral responses.
Collapse
Affiliation(s)
- Luis M Molinos-Albert
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.
| | - Rocío Rubio
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Carla Martín-Pérez
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Edwards Pradenas
- IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona (Barcelona), Spain
| | - Cèlia Torres
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Alfons Jiménez
- ISGlobal, Barcelona, Spain; CIBER de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Mar Canyelles
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Marta Vidal
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | | | - Silvia Marfil
- IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona (Barcelona), Spain
| | - Ester Aparicio
- IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona (Barcelona), Spain
| | - Anna Ramírez-Morros
- Unitat de Suport a la Recerca de la Catalunya Central, Fundació Institut Universitari per a la Recerca a l'Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJG), Manresa, Spain
| | - Benjamin Trinité
- IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona (Barcelona), Spain
| | - Josep Vidal-Alaball
- Unitat de Suport a la Recerca de la Catalunya Central, Fundació Institut Universitari per a la Recerca a l'Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJG), Manresa, Spain; Health Promotion in Rural Areas Research Group (PROSAARU), Gerència Territorial de la Catalunya Central, Institut Català de la Salut, Manresa, Spain; Universitat de Vic-Universitat Central de Catalunya (uVic-UCC), Vic, Spain
| | - Pere Santamaria
- Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain; Department of Microbiology, Immunology, and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Pau Serra
- Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain
| | - Luis Izquierdo
- ISGlobal, Barcelona, Spain; CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Ruth Aguilar
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Anna Ruiz-Comellas
- Unitat de Suport a la Recerca de la Catalunya Central, Fundació Institut Universitari per a la Recerca a l'Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJG), Manresa, Spain; Health Promotion in Rural Areas Research Group (PROSAARU), Gerència Territorial de la Catalunya Central, Institut Català de la Salut, Manresa, Spain; Centre d'Atenció Primària (CAP) Sant Joan de Vilatorrada, Gerència Territorial de la Catalunya Central, Institut Català de la Salut (ICS), Manresa, Spain; Universitat de Vic-Universitat Central de Catalunya (uVic-UCC), Vic, Spain
| | - Julià Blanco
- IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona (Barcelona), Spain; CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain; Universitat de Vic-Universitat Central de Catalunya (uVic-UCC), Vic, Spain; Germans Trias i Pujol Research Institute (IGTP), Campus Can Ruti, Badalona (Barcelona), Spain
| | - Carlota Dobaño
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain; CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.
| | - Gemma Moncunill
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain; CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
16
|
Bachiller S, Vitallé J, Camprubí-Ferrer L, García M, Gallego I, López-García M, Galvá MI, Cañizares J, Rivas-Jeremías I, Díaz-Mateos M, Gasca-Capote C, Moral-Turón C, Galán-Villamor L, Fontillón M, Sobrino S, Cisneros JM, López-Cortés LF, Deierborg T, Ruiz-Mateos E. SARS-CoV-2 post-acute sequelae linked to inflammation via extracellular vesicles. Front Immunol 2025; 16:1501666. [PMID: 40330474 PMCID: PMC12052859 DOI: 10.3389/fimmu.2025.1501666] [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: 10/07/2024] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
Background Despite the efficacy of SARS-CoV-2 vaccines in reducing mortality and severe cases of COVID-19, a proportion of survivors experience long-term symptoms, known as post-acute sequelae of SARS-CoV-2 infection (PASC). This study investigates the long-term immunological and neurodegenerative effects associated with extracellular vesicles (EVs) in COVID-19 survivors, 15 months after SARS-CoV-2 infection. Methods 13 Controls and 20 COVID-19 survivors, 15 months after SARS-CoV-2 infection, were recruited. Pro-inflammatory cytokines were analyzed in both plasma and EVs. A deep-immunophenotyping of monocytes, T-cells and dendritic cells (DCs) was performed, along with immunostainings of SARS-CoV-2 in the colon. Results Higher concentrations of pro-inflammatory cytokines and neurofilaments were found in EVs but not in plasma from COVID-19 survivors. Additionally, COVID-19 participants exhibited altered monocyte activation markers and elevated cytokine production upon lipopolysaccharide stimulation. Increased activation markers in CD4+ T-cells and decreased indoleamine 2,3-dioxygenase expression in DCs were observed in COVID-19 participants. Furthermore, the amount of plasmacytoid DCs expressing β7-integrin were higher in COVID-19, potentially associated with the viral persistence observed in the colon. Conclusions COVID-19 survivors exhibit long-term immune dysregulation and neurodegeneration, emphasizing the need for ongoing monitoring of PASC. The cargo of EVs can be a promising tool for early detection of virus-induced neurological disorders.
Collapse
Affiliation(s)
- Sara Bachiller
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital, Spanish National Research Council (CSIC), University of Seville, Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Seville, Spain
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Joana Vitallé
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital, Spanish National Research Council (CSIC), University of Seville, Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Seville, Spain
| | - Lluís Camprubí-Ferrer
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Manuel García
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital, Spanish National Research Council (CSIC), University of Seville, Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Seville, Spain
| | - Isabel Gallego
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital, Spanish National Research Council (CSIC), University of Seville, Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Seville, Spain
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | | | | | | | - Inmaculada Rivas-Jeremías
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital, Spanish National Research Council (CSIC), University of Seville, Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Seville, Spain
| | | | - Carmen Gasca-Capote
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital, Spanish National Research Council (CSIC), University of Seville, Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Seville, Spain
| | - Cristina Moral-Turón
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital, Spanish National Research Council (CSIC), University of Seville, Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Seville, Spain
| | | | - María Fontillón
- Service of Pathological Anatomy, Virgen del Rocío University Hospital, Seville, Spain
| | - Salvador Sobrino
- Digestive Service, Virgen del Rocío University Hospital, Seville, Spain
| | - José Miguel Cisneros
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital, Spanish National Research Council (CSIC), University of Seville, Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Seville, Spain
| | - Luis Fernando López-Cortés
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital, Spanish National Research Council (CSIC), University of Seville, Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Seville, Spain
| | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Ezequiel Ruiz-Mateos
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital, Spanish National Research Council (CSIC), University of Seville, Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Seville, Spain
| |
Collapse
|
17
|
Karl V, Hofmann M, Thimme R. Role of antiviral CD8+ T cell immunity to SARS-CoV-2 infection and vaccination. J Virol 2025; 99:e0135024. [PMID: 40029063 PMCID: PMC11998524 DOI: 10.1128/jvi.01350-24] [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] [Indexed: 03/05/2025] Open
Abstract
The COVID-19 pandemic has greatly enhanced our understanding of CD8+ T cell immunity and their role in natural infection and vaccine-induced protection. Rapid and early SARS-CoV-2-specific CD8+ T cell responses have been associated with efficient viral clearance and mild disease. Virus-specific CD8+ T cell responses can compensate for waning, morbidity-related, and iatrogenic reduction of humoral immunity. After infection or vaccination, SARS-CoV-2-specific memory CD8+ T cells are formed, which mount an efficient recall response in the event of breakthrough infection and help to protect from severe disease. Due to their breadth and ability to target mainly highly conserved epitopes, SARS-CoV-2-specific CD8+ T cells are also able to cross-recognize epitopes of viral variants, thus maintaining immunity even after the emergence of viral evolution. In some cases, however, CD8+ T cells may contribute to the pathogenesis of severe COVID-19. In particular, delayed and uncontrolled, e.g., nonspecific and hyperactivated, cytotoxic CD8+ T cell responses have been linked to poor COVID-19 outcomes. In this minireview, we summarize the tremendous knowledge about CD8+ T cell responses to SARS-CoV-2 infection and COVID-19 vaccination that has been gained over the past 5 years, while also highlighting the critical knowledge gaps that remain.
Collapse
Affiliation(s)
- Vivien Karl
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Maike Hofmann
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Thimme
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
18
|
Pernet O, Frederick T, Adili A, Hudgins J, Anthony P, McCaney G, Mack WJ, Noriega E, Lopez J, Balog S, Biniwale M, Yeh A, Bearden A, Ramanathan R, Kovacs A. Cord blood IgA/M reveals in utero response to SARS-CoV-2 with fluctuations in relation to circulating variants. Nat Commun 2025; 16:3551. [PMID: 40229328 PMCID: PMC11997084 DOI: 10.1038/s41467-025-58768-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 04/02/2025] [Indexed: 04/16/2025] Open
Abstract
It is estimated that in utero SARS-CoV-2 infection is rare. However, few studies have systematically assessed for IgA and IgM antibodies indicating potential in utero response to SARS-CoV-2 infection using multi-isotype serology, and no studies have assessed in utero infection markers in relation to circulating variants. Between October 21, 2021 and February 15, 2023, remnant cord blood samples (CBS) from neonates born at a single hospital in Los Angeles, were systematically tested for serological markers suggesting in utero infection. SARS-CoV-2 specific fetal IgA and/or IgM antibodies were detected in 28.7% (298/1038 CBS, 95% CI: 26.0, 31.6), higher than previous in utero infection estimates that used only PCR and/or IgM. Importantly, the probability of detecting markers of in utero infection varied by month (P-value = 0.0144). The prevalence of fetal IgA/IgM varied with the emergence of new variants, increasing during the BA.1 wave with a peak in February 2022 at 36% (18/50, 95% CI: 22.7-49.3) and again during the BA.4/5 wave, with a peak at 48.8% in September 2022 (39/80, 95% CI 37.8-59.7), suggesting variant-related fluctuations. These data suggest it may be useful to identify SARS-Cov-2 in utero exposure at birth so these newborns may be more closely followed for adverse clinical outcomes.
Collapse
Affiliation(s)
- Olivier Pernet
- Maternal, Child, and Adolescent Center for Infectious Diseases and Virology, Division of Pediatric Infectious Diseases, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA.
| | - Toinette Frederick
- Maternal, Child, and Adolescent Center for Infectious Diseases and Virology, Division of Pediatric Infectious Diseases, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA
| | - Amila Adili
- Southern California Clinical and Translational Science Institute (SC-CTSI), University of Southern California, Los Angeles, CA, USA
| | - Jay Hudgins
- Department of Pathology and Laboratory Medicine, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA
| | - Patricia Anthony
- Maternal, Child, and Adolescent Center for Infectious Diseases and Virology, Division of Pediatric Infectious Diseases, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA
| | - Gwyndolyn McCaney
- Maternal, Child, and Adolescent Center for Infectious Diseases and Virology, Division of Pediatric Infectious Diseases, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA
| | - Wendy J Mack
- Southern California Clinical and Translational Science Institute (SC-CTSI), University of Southern California, Los Angeles, CA, USA
- Department of Population and Public Health Sciences, University of Southern California, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Eunice Noriega
- Maternal, Child, and Adolescent Center for Infectious Diseases and Virology, Division of Pediatric Infectious Diseases, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA
| | - Jennifer Lopez
- Maternal, Child, and Adolescent Center for Infectious Diseases and Virology, Division of Pediatric Infectious Diseases, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA
| | - Steven Balog
- Maternal, Child, and Adolescent Center for Infectious Diseases and Virology, Division of Pediatric Infectious Diseases, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA
| | - Manoj Biniwale
- Division of Neonatal Medicine, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA
| | - Amy Yeh
- Division of Neonatal Medicine, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA
| | - Allison Bearden
- Maternal, Child, and Adolescent Center for Infectious Diseases and Virology, Division of Pediatric Infectious Diseases, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA
| | - Rangasamy Ramanathan
- Division of Neonatal Medicine, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA
| | - Andrea Kovacs
- Maternal, Child, and Adolescent Center for Infectious Diseases and Virology, Division of Pediatric Infectious Diseases, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA.
- Department of Pathology and Laboratory Medicine, University of Southern California, Keck School of Medicine of USC, and Los Angeles General Medical Center, Los Angeles, CA, USA.
- Department of Population and Public Health Sciences, University of Southern California, Keck School of Medicine of USC, Los Angeles, CA, USA.
| |
Collapse
|
19
|
Carter JY, Khamadi S, Mwangi J, Muhula S, Munene SM, Kanyara L, Kinyua J, Lagat N, Chege J, Oira R, Maiyo A, Stewart R, Postma M, Stekelenburg J, Osur J, van Hulst M. Seroprevalence and demographic characteristics of SARS-CoV-2-infected residents of Kibera informal settlement during the COVID-19 pandemic in Nairobi, Kenya: a cross-sectional study. BMJ Open 2025; 15:e094546. [PMID: 40180388 PMCID: PMC11966967 DOI: 10.1136/bmjopen-2024-094546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 03/07/2025] [Indexed: 04/05/2025] Open
Abstract
OBJECTIVES To assess the prevalence of SARS-CoV-2 antibodies in the residents of Kibera informal settlement in Nairobi, Kenya, before vaccination became widespread, and explore demographic and health-related risk factors for infection. DESIGN A cross-sectional study. SETTING Kibera informal settlement, Nairobi, Kenya. PARTICIPANTS Residents of Kibera informal settlement between October 2019 and August 2021, age 1 year and above who reported no current symptoms of COVID-19. MAIN OUTCOME MEASURES Associations were determined between SARS-CoV-2 positive tests measured with one rapid test and two ELISAs and demographic and health-related factors, using Pearson's χ2 test. Crude OR and adjusted OR were calculated to quantify the strength of associations between variables and seropositive status. RESULTS A total of 438 participants were recruited. Most (79.2%) were age 18-50 years; females (64.2%) exceeded males. More than one-third (39.1%) were unemployed; only 7.4% were in formal, full-time employment. Less than one-quarter (22.1%) self-reported any underlying health conditions. Nearly two-thirds (64.2%) reported symptoms compatible with COVID-19 in the previous 16 months; only one (0.23%) had been hospitalised with a reported negative COVID-19 test. 370 (84.5%) participants tested positive in any of the three tests. There was no significant difference in SARS-CoV-2 seropositivity across age, sex, presence of underlying health conditions, on medication or those ever tested for SARS-CoV-2. Multiple logistic regression analysis showed that COVID-19 symptoms in the previous 16 months were the only significant independent predictor of seropositivity (p=0.0085). CONCLUSION High SARS-CoV-2 exposure with limited morbidity was found in the residents of Kibera informal settlement. The study confirms other reports of high SARS-CoV-2 exposure with limited morbidity in slum communities. Reasons cited include the high infectious disease burden on the African continent, demographic age structure and underreporting due to limited testing and lack of access to healthcare services; genetic factors may also play a role. These factors require further investigation.
Collapse
Affiliation(s)
- Jane Y Carter
- Regional Laboratory Programme, Amref Health Africa, Nairobi, Kenya
| | - Samoel Khamadi
- Centre for Virus Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Joseph Mwangi
- Centre for Virus Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Samuel Muhula
- Monitoring, Evaluation and Learning Department, Amref Health Africa, Nairobi, Kenya
| | - Stephen M Munene
- Regional Laboratory Programme, Amref Health Africa, Nairobi, Kenya
| | - Lucy Kanyara
- Centre for Virus Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Joyceline Kinyua
- Centre for Virus Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Nancy Lagat
- Centre for Virus Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Judy Chege
- Centre for Virus Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Robert Oira
- Centre for Virus Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Alex Maiyo
- Centre for Virus Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Roy Stewart
- Department of Health Sciences, Community and Occupational Medicine, University Medical Centre Groningen, Groningen, The Netherlands
| | - Maarten Postma
- Department of Health Sciences, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
- Department of Economics, Econometrics and Finance, University of Groningen, Faculty of Economics and Business, Groningen, The Netherlands
- Center of Excellence for Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia
- Division of Pharmacology and Therapy, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Jelle Stekelenburg
- Department of Health Sciences, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
- Department of Obstetrics and Gynaecology, Frisius Medical Center Leeuwarden, Leeuwarden, The Netherlands
| | - Joachim Osur
- Office of the Vice Chancellor, Amref International University, Nairobi, Kenya
| | - Marinus van Hulst
- Department of Health Sciences, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
- Department of Clinical Pharmacy and Toxicology, Martini Hospital, Groningen, The Netherlands
| |
Collapse
|
20
|
Markey G, McLaughlin J, McDaid D, Lynch SM, English A, Alexander HD, Kelly M, Bhavsar M, McGilligan V, Zhang SD, Murray EK, Rai TS, Walsh C, Bjourson AJ, Shukla P, Gibson DS. Distinct Omicron longitudinal memory T cell profile and T cell receptor repertoire associated with COVID-19 hospitalisation. Front Immunol 2025; 16:1549570. [PMID: 40242761 PMCID: PMC12000046 DOI: 10.3389/fimmu.2025.1549570] [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: 12/21/2024] [Accepted: 02/20/2025] [Indexed: 04/18/2025] Open
Abstract
SARS-CoV-2 has claimed more than 7 million lives worldwide and has been associated with prolonged inflammation, immune dysregulation and persistence of symptoms following severe infection. Understanding the T cell mediated immune response and factors impacting development and continuity of SARS-CoV-2 specific memory T cells is pivotal for developing better therapeutic and monitoring strategies for those most at risk from COVID-19. Here we present a comprehensive analysis of memory T cells in a convalescent cohort (n=20), three months post Omicron infection. Utilising flow cytometry to investigate CD4+CD45RO+ and CD8+CD45RO+ memory T cell IL-2 expression following Omicron (B.1.1.529/BA.1) peptide pool stimulation, alongside T cell receptor repertoire profiling and RNA-Seq analysis, we have identified several immunological features associated with hospitalised status. We observed that while there was no significant difference in median CD4+CD45RO+ IL-2+ and CD8+ CD45RO+ IL-2+ memory T cell count between subgroups, the hospitalised subgroup expressed significantly more IL-2 per cell following Omicron peptide pool exposure in the CD8+CD45RO+ population (p <0.03) and trended towards significance in CD4+CD45RO+ cells (p <0.06). T cell receptor repertoire analysis found that the non-hospitalised subgroup had a much higher number of circulating clonotypes, targeting a wider range of predominantly MHC-I epitopes across the SARS-CoV-2 genome. Several immunodominant epitopes, conserved between both subgroups, were observed, however hospitalised individuals were less likely to express putative HLA alleles responsible for pMHC presentation which may impact TCR affinity. We observed a bias towards shorter CDR3 segments in TCRβ repertoire analysis within the hospitalised subgroup, alongside lower rates of repertoire overlap in CDR3 sequences compared to the non-hospitalised subgroup. We found a significant proportion of TCRs targeted epitopes along the SARS-CoV-2 genome including non-structural proteins, responsible for viral replication and immune evasion. These findings highlight how the continuity of T cell based protective immunity is impacted by both the viral replication cycle of SARS-CoV-2 upon intracellular and innate immune responses, and HLA-type upon TCR affinity and clonotype formation. Our novel Epitope Target Analysis Pipeline (Epi-TAP) could prove beneficial in development of new therapeutic strategies through rapid identification of shared immunodominant epitopes across non-hospitalised and hospitalised subgroups.
Collapse
Affiliation(s)
- Gavin Markey
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry, United Kingdom
| | - Joseph McLaughlin
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry, United Kingdom
- Medical Directorate, Clinical and Translational Research and Innovation Centre, Londonderry, United Kingdom
| | - Darren McDaid
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry, United Kingdom
| | - Seodhna M. Lynch
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry, United Kingdom
| | - Andrew English
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry, United Kingdom
- School of Health and Life Sciences, Teesside University, Middlesborough, United Kingdom
| | - H. Denis Alexander
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry, United Kingdom
| | - Martin Kelly
- Intensive Care Unit, Western Health Social Care Trust, Londonderry, United Kingdom
| | - Manav Bhavsar
- Intensive Care Unit, Western Health Social Care Trust, Londonderry, United Kingdom
| | - Victoria McGilligan
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry, United Kingdom
| | - Shu-Dong Zhang
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry, United Kingdom
| | - Elaine K. Murray
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry, United Kingdom
| | - Taranjit Singh Rai
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry, United Kingdom
| | - Colum Walsh
- Genomic Medicine Research Group, School of Biomedical Science, Ulster University, Coleraine, United Kingdom
- Biomedical and Clinical Sciences Division, Department for Cell and Neurobiology, Faculty of Medicine, Linköping University, Linköping, Sweden
| | - Anthony J. Bjourson
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry, United Kingdom
| | - Priyank Shukla
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry, United Kingdom
| | - David S. Gibson
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry, United Kingdom
| |
Collapse
|
21
|
Benlarbi M, Kenfack DD, Dionne K, Côté-Chenette M, Beaudoin-Bussières G, Bélanger É, Ding S, Goni OH, Ngoume YF, Tauzin A, Medjahed H, Ghedin E, Duerr R, Finzi A, Tongo M. Longitudinal humoral immunity against SARS-CoV-2 Spike following infection in individuals from Cameroon. Virology 2025; 605:110467. [PMID: 40037139 PMCID: PMC11937844 DOI: 10.1016/j.virol.2025.110467] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/10/2025] [Accepted: 02/24/2025] [Indexed: 03/06/2025]
Abstract
In May 2023 the World Health Organization (WHO) declared the end of COVID-19 as a public health emergency. Seroprevalence studies performed in African countries, such as Cameroon, depicted a much higher COVID-19 burden than reported by the WHO. To better understand humoral responses kinetics following infection, we enrolled 333 participants from Yaoundé, Cameroon between March 2020 and January 2022. We measured the levels of antibodies targeting the SARS-CoV-2 receptor-binding-domain (RBD) and the Spike glycoproteins of Delta, Omicron BA.1 and BA.4/5 and the common cold coronavirus HCoV-OC43. We also evaluated plasma capacity to neutralize authentic SARS-CoV-2 virus and to mediate Antibody-Dependent Cellular Cytotoxicity (ADCC). Most individuals mounted a strong antibody response against SARS-CoV-2 Spike. Plasma neutralization waned faster than anti-Spike binding and ADCC. We observed differences in humoral responses by age and circulating variants. Altogether, we show a global overview of antibody dynamics and functionality against SARS-CoV-2 in Cameroon.
Collapse
Affiliation(s)
- Mehdi Benlarbi
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Dell-Dylan Kenfack
- Center of Research for Emerging and Re-Emerging Diseases (CREMER), Institute of Medical Research and Study of Medicinal Plants (IMPM), Yaoundé, Cameroon
| | - Katrina Dionne
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Maxime Côté-Chenette
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Guillaume Beaudoin-Bussières
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Étienne Bélanger
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Shilei Ding
- Centre de Recherche du CHUM, Montréal, Québec, Canada
| | - Oumarou H Goni
- Center of Research for Emerging and Re-Emerging Diseases (CREMER), Institute of Medical Research and Study of Medicinal Plants (IMPM), Yaoundé, Cameroon
| | - Yannick F Ngoume
- Center of Research for Emerging and Re-Emerging Diseases (CREMER), Institute of Medical Research and Study of Medicinal Plants (IMPM), Yaoundé, Cameroon
| | - Alexandra Tauzin
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Halima Medjahed
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Elodie Ghedin
- Systems Genomics Section, Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, Bethesda, MD, USA
| | - Ralf Duerr
- Vaccine Center, NYU Grossman School of Medicine, New York, USA; Department of Medicine, NYU Grossman School of Medicine, New York, USA; Department of Microbiology, NYU Grossman School of Medicine, New York, USA
| | - Andrés Finzi
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada.
| | - Marcel Tongo
- Center of Research for Emerging and Re-Emerging Diseases (CREMER), Institute of Medical Research and Study of Medicinal Plants (IMPM), Yaoundé, Cameroon; HIV Pathogenesis Program, The Doris Duke Medical Research Institute, University of KwaZulu Natal, Durban, South Africa.
| |
Collapse
|
22
|
Case JB, Jain S, Suthar MS, Diamond MS. SARS-CoV-2: The Interplay Between Evolution and Host Immunity. Annu Rev Immunol 2025; 43:29-55. [PMID: 39705164 DOI: 10.1146/annurev-immunol-083122-043054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2024]
Abstract
The persistence of SARS-CoV-2 infections at a global level reflects the repeated emergence of variant strains encoding unique constellations of mutations. These variants have been generated principally because of a dynamic host immune landscape, the countermeasures deployed to combat disease, and selection for enhanced infection of the upper airway and respiratory transmission. The resulting viral diversity creates a challenge for vaccination efforts to maintain efficacy, especially regarding humoral aspects of protection. Here, we review our understanding of how SARS-CoV-2 has evolved during the pandemic, the immune mechanisms that confer protection, and the impact viral evolution has had on transmissibility and adaptive immunity elicited by natural infection and/or vaccination. Evidence suggests that SARS-CoV-2 evolution initially selected variants with increased transmissibility but currently is driven by immune escape. The virus likely will continue to drift to maintain fitness until countermeasures capable of disrupting transmission cycles become widely available.
Collapse
Affiliation(s)
- James Brett Case
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA;
| | - Shilpi Jain
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mehul S Suthar
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michael S Diamond
- Department of Pathology & Immunology; Department of Molecular Microbiology; and Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA;
| |
Collapse
|
23
|
Yuan L, Stoddard M, Sarkar S, van Egeren D, Mangalaganesh S, Nolan RP, Rogers MS, Hather G, White LF, Chakravarty A. The Impact of Vaccination Frequency on COVID-19 Public Health Outcomes: A Model-Based Analysis. Vaccines (Basel) 2025; 13:368. [PMID: 40333247 PMCID: PMC12031506 DOI: 10.3390/vaccines13040368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 03/16/2025] [Accepted: 03/27/2025] [Indexed: 05/09/2025] Open
Abstract
Background: While the rapid deployment of SARS-CoV-2 vaccines had a significant impact on the ongoing COVID-19 pandemic, rapid viral immune evasion and waning neutralizing antibody titers have degraded vaccine efficacy. Nevertheless, vaccine manufacturers and public health authorities have a number of options at their disposal to maximize the benefits of vaccination. In particular, the effect of booster schedules on vaccine performance bears further study. Methods: To better understand the effect of booster schedules on vaccine performance, we used an agent-based modeling framework and a population pharmacokinetic model to simulate the impact of boosting frequency on the durability of vaccine protection against infection and severe acute disease. Results: Our work suggests that repeated dosing at frequent intervals (three or more times a year) may offset the degradation of vaccine efficacy, preserving the utility of vaccines in managing the ongoing pandemic. Conclusions: Given the practical significance of potential improvements in vaccine utility, clinical research to better understand the effects of repeated vaccination would be highly impactful. These findings are particularly relevant as public health authorities worldwide have reduced the frequency of boosters to once a year or less.
Collapse
Affiliation(s)
- Lin Yuan
- Fractal Therapeutics, Lexington, MA 02420, USA; (L.Y.); (M.S.)
| | | | - Sharanya Sarkar
- Department of Microbiology and Immunology, Dartmouth College, Hanover, NH 03755, USA;
| | - Debra van Egeren
- Department of Oncology, School of Medicine, Stanford University, Stanford, CA 94305, USA;
| | - Shruthi Mangalaganesh
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC 3800, Australia;
| | | | - Michael S. Rogers
- Department of Surgery, Harvard Medical School, Boston, MA 02114, USA;
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Greg Hather
- Sage Therapeutics, Cambridge, MA 02142, USA;
| | - Laura F. White
- School of Public Health, Boston University, Boston, MA 02118, USA;
| | | |
Collapse
|
24
|
Ahmadivand S, Fux R, Palić D. Role of T Follicular Helper Cells in Viral Infections and Vaccine Design. Cells 2025; 14:508. [PMID: 40214462 PMCID: PMC11987902 DOI: 10.3390/cells14070508] [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: 01/25/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025] Open
Abstract
T follicular helper (Tfh) cells are a specialized subset of CD4+ T lymphocytes that are essential for the development of long-lasting humoral immunity. Tfh cells facilitate B lymphocyte maturation, promote germinal center formation, and drive high-affinity antibody production. Our current knowledge of Tfh interactions with the humoral immune system effectors suggests that they have a critical role in supporting the immune response against viral infections. This review discusses the mechanisms through which Tfh cells influence anti-viral immunity, highlighting their interactions with B cells and their impact on antibody quality and quantity. We explore the role of Tfh cells in viral infections and examine how vaccine design can be improved to enhance Tfh cell responses. Innovative vaccine platforms, such as mRNA vaccines and self-assembling protein nanoplatforms (SAPNs), are promising strategies to enhance Tfh cell activation. Their integration and synergistic combination could further enhance immunity and Tfh responses (SAPN-RNA vaccines). In summary, we provide a comprehensive overview of the current insights into Tfh cells' role during viral infections, emphasizing their potential as strategic targets for innovative vaccine development.
Collapse
Affiliation(s)
- Sohrab Ahmadivand
- Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, 80539 Munich, Germany
| | - Robert Fux
- Institute for Infectious Diseases and Zoonoses, Ludwig-Maximilians-University Munich, 80539 Munich, Germany;
| | - Dušan Palić
- Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, 80539 Munich, Germany
| |
Collapse
|
25
|
Hsu CY, Chiu CH, Lin TY, Chang FY. Severe SARS-CoV-2 alpha variant convalescent patients exhibit worse T cell immune response than those with mild severity disease. J Formos Med Assoc 2025:S0929-6646(25)00110-X. [PMID: 40155285 DOI: 10.1016/j.jfma.2025.03.009] [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: 10/04/2024] [Revised: 01/14/2025] [Accepted: 03/07/2025] [Indexed: 04/01/2025] Open
Abstract
OBJECTIVES This study aimed to assess T cell phenotype and the role of cellular immunity against acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants in alpha variant coronavirus disease 2019 (COVID-19) convalescent patients. METHODS Thirty-two confirmed SARS-CoV-2 infected patients and ten healthy controls were enrolled. T cell subsets in peripheral blood were classified and quantified using flow cytometry. Additionally, T cell immune responses against SARS-CoV-2 spike peptide pools were assessed. Flow cytometry data were analyzed using Cytobank software. Other analyses involved Student's t-test or chi-square test. RESULTS CD127 expression on T helper cells and cytotoxic T cells was lower in the severe disease group than that in the mild disease group. Severe COVID-19 convalescents with SARS-CoV-2 alpha variant exhibited poorer T cell immune responses than those with mild disease upon spike peptide pools stimulation with SARS-CoV-2 wild type, alpha, or omicron variants. CONCLUSIONS COVID-19 convalescents showed T helper and cytotoxic T cells with lower CD127 expression in the severe disease group than those in the mild disease group. Severe COVID-19 convalescents infected with the alpha variant exhibited poorer T cell immune responses against the SARS-CoV-2 wild type, alpha, or omicron variants. Our study provides insights into the differences in T cell phenotypes and immune responses during the contraction phase following SARS-CoV-2 infection across varying disease severities. These findings offer valuable perspectives for advancing future research on SARS-CoV-2 T cell-related immune responses.
Collapse
Affiliation(s)
- Chih-Yao Hsu
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chun-Hsiang Chiu
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Te-Yu Lin
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| | - Feng-Yee Chang
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
26
|
Florian DM, Bauer M, Popovitsch A, Fae I, Springer DN, Graninger M, Traugott M, Weseslindtner L, Aberle SW, Fischer G, Kundi M, Stiasny K, Zoufaly A, Landry SJ, Aberle JH. Enhanced and long-lasting SARS-CoV-2 immune memory in individuals with common cold coronavirus cross-reactive T cell immunity. Front Immunol 2025; 16:1501704. [PMID: 40191213 PMCID: PMC11968687 DOI: 10.3389/fimmu.2025.1501704] [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: 09/25/2024] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
With the continuous emergence of novel SARS-CoV-2 variants, long-lasting and broadly reactive cellular and humoral immunity is critical for durable protection from COVID-19. We investigated SARS-CoV-2-specific T cell immunity in relation to antibodies, infection outcome and disease severity and assessed its durability in a longitudinal cohort over a three-year time course. We identified pre-existing T cells reactive to the seasonal coronavirus (CoV) OC43 that cross-react with the conserved SARS-CoV-2 spike S813-829 peptide. These cross-reactive T cells increased in frequency following SARS-CoV-2 infection or vaccination and correlated with enhanced spike-specific T cell responses and significantly reduced viral loads. Furthermore, our data revealed that CoV-cross-reactive T cells were maintained as part of the long-lasting memory response, contributing to increased T cell frequencies against omicron variants. These findings suggest a functional role of CoV-cross-reactive T cells that extends beyond the initial SARS-CoV-2 exposure, contributing to enhanced immunity against highly mutated SARS-CoV-2 variants.
Collapse
Affiliation(s)
- David M. Florian
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Michael Bauer
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | | | - Ingrid Fae
- Department of Transfusion Medicine and Cell Therapy, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | - Gottfried Fischer
- Department of Transfusion Medicine and Cell Therapy, Medical University of Vienna, Vienna, Austria
| | - Michael Kundi
- Center for Public Health, Department for Environmental Health, Medical University of Vienna, Vienna, Austria
| | - Karin Stiasny
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Alexander Zoufaly
- Department of Medicine IV, Klinik Favoriten, Vienna, Austria
- Faculty of Medicine, Sigmund Freud University, Vienna, Austria
| | - Samuel J. Landry
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Judith H. Aberle
- Center for Virology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
27
|
Piano Mortari E, Ferrucci F, Zografaki I, Carsetti R, Pacelli L. T and B cell responses in different immunization scenarios for COVID-19: a narrative review. Front Immunol 2025; 16:1535014. [PMID: 40170841 PMCID: PMC11959168 DOI: 10.3389/fimmu.2025.1535014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/25/2025] [Indexed: 04/03/2025] Open
Abstract
Vaccines against COVID-19 have high efficacy and low rates of adverse events. However, none of the available vaccines provide sterilizing immunity, and reinfections remain possible. This review aims to summarize the immunological responses elicited by different immunization strategies, examining the roles of homologous and heterologous vaccination and hybrid immunity. Homologous vaccination regimens exhibit considerable variation in immune responses depending on the vaccine platform, particularly concerning antibody titers, B cell activation, and T cell responses. mRNA vaccines, such as mRNA-1273 and BNT162b2, consistently generate higher and more durable levels of neutralizing antibodies and memory B cells compared to adenovirus-based vaccines like Ad26.COV2.S and ChAdOx1. The combination of two distinct vaccine platforms, each targeting different immune pathways, seems to be more effective in promoting long-lasting B cell responses and potent T cell responses. The high heterogeneity of the available studies, the different dosing schemes, the succession of new variants, and the subjects' immunological background do not allow for a definitive conclusion. Overall, heterologous vaccination strategies, combining sequentially viral vector and mRNA may deliver a more balanced and robust humoral and cellular immune response compared to homologous regimens. Hybrid immunity, which arises from SARS-CoV-2 infection preceded or followed by vaccination produces markedly stronger immune responses than either vaccination or infection alone. The immune response to SARS-CoV-2 variants of concern varies depending on both the vaccine platform and prior infection status. Hybrid immunity leads to a broader antibody repertoire, providing enhanced neutralization of variants of concern. Heterologous vaccination and hybrid immunity may provide further opportunities to enhance immune responses, offering broader protection and greater durability of immunity. However, from all-cause mortality, symptomatic or severe COVID, and serious adverse events at present it is not possible to infer different effects between homologous and heterologous schemes. Next-generation vaccines could involve tweaks to these designs or changes to delivery mechanisms that might improve performance.
Collapse
Affiliation(s)
- Eva Piano Mortari
- B Lymphocytes Unit, Bambino Gesù Children’s Hospital, istituto di ricovero e cura a carattere scientifico (IRCCS), Rome, Italy
| | | | - Irini Zografaki
- mRNA & Antivirals Medical & Scientific Affairs International Developed Markets, Pfizer, Athens, Greece
| | - Rita Carsetti
- B Lymphocytes Unit, Bambino Gesù Children’s Hospital, istituto di ricovero e cura a carattere scientifico (IRCCS), Rome, Italy
| | - Luciano Pacelli
- Medical Department, Internal Medicine, Pfizer s.r.l., Rome, Italy
| |
Collapse
|
28
|
Samaan P, Korosec CS, Budylowski P, Chau SLL, Pasculescu A, Qi F, Delgado-Brand M, Tursun TR, Mailhot G, Dayam RM, Arnold CR, Langlois MA, Mendoza J, Morningstar T, Law R, Mihelic E, Sheikh-Mohamed S, Cao EY, Paul N, Patel A, de Launay KQ, Boyd JM, Takaoka A, Colwill K, Matveev V, Yue FY, McGeer A, Straus S, Gingras AC, Heffernen JM, Ostrowski M. mRNA vaccine-induced SARS-CoV-2 spike-specific IFN-γ and IL-2 T-cell responses are predictive of serological neutralization and are transiently enhanced by pre-existing cross-reactive immunity. J Virol 2025; 99:e0168524. [PMID: 39887249 PMCID: PMC11915849 DOI: 10.1128/jvi.01685-24] [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: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 02/01/2025] Open
Abstract
The contributions of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific T cells to vaccine efficacy and durability are unclear. We investigated relationships between mRNA vaccine-induced spike-specific interferon- gamma (IFN-γ) and interleukin-2 (IL-2) T-cell responses and neutralizing antibody development in long-term care home staff doubly vaccinated with BNT162b2 or mRNA-1273. The impacts of pre-existing cross-reactive T-cell immunity on cellular and humoral responses to vaccination were additionally assessed. Mathematical modeling of the kinetics of spike-specific IFN-γ and IL-2 T-cell responses over 6 months post-second dose was bifurcated into recipients who exhibited gradual increases with doubling times of 155 and 167 days or decreases with half-lives of 165 and 132 days, respectively. Differences in kinetics did not correlate with clinical phenotypes. Serological anti-spike IgG, anti-receptor binding domain (RBD) IgG, anti-spike IgA, and anti-RBD IgA antibody levels otherwise decayed in all participants with half-lives of 63, 57, 79, and 46 days, respectively, alongside waning neutralizing capacity (t1/2 = 408 days). Spike-specific T-cell responses induced at 2-6 weeks positively correlated with live viral neutralization at 6 months post-second dose, especially in hybrid immune individuals. Participants with pre-existing cross-reactive T-cell immunity to SARS-CoV-2 exhibited greater spike-specific T-cell responses, reduced anti-RBD IgA antibody levels, and a trending increase in neutralization at 2-6 weeks post-second dose. Non-spike-specific T-cells predominantly targeted SARS-CoV-2 non-structural protein at 6 months post-second dose in cross-reactive participants. mRNA vaccination was lastly shown to induce off-target T-cell responses against unrelated antigens. In summary, vaccine-induced spike-specific T-cell immunity appeared to influence serological neutralizing capacity, with only a modest effect induced by pre-existing cross-reactivity. IMPORTANCE Our findings provide valuable insights into the potential contributions of mRNA vaccine-induced spike-specific T-cell responses to the durability of neutralizing antibody levels in both uninfected and hybrid immune recipients. Our study additionally sheds light on the precise impacts of pre-existing cross-reactive T-cell immunity to severe acute respiratory syndrome coronavirus 2 on the magnitude and kinetics of cellular and humoral responses to vaccination. Accordingly, our data will help optimize the development of next-generation T cell-based coronavirus vaccines and vaccine regimens to maximize efficacy and durability.
Collapse
Affiliation(s)
- Philip Samaan
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Chapin S. Korosec
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, Toronto, Ontario, Canada
- Center for Disease Modelling, Mathematics and Statistics, York University, Toronto, Ontario, Canada
| | - Patrick Budylowski
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Serena L. L. Chau
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Adrian Pasculescu
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Freda Qi
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | | | - Tulunay R. Tursun
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Geneviève Mailhot
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Roya Monica Dayam
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Corey R. Arnold
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Justin Mendoza
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Ryan Law
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Erik Mihelic
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Eric Yixiao Cao
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Nimitha Paul
- Unity Health Toronto, St Michael's Hospital, Toronto, Ontario, Canada
| | - Anjali Patel
- Unity Health Toronto, St Michael's Hospital, Toronto, Ontario, Canada
| | | | - Jamie M. Boyd
- Unity Health Toronto, St Michael's Hospital, Toronto, Ontario, Canada
| | - Alyson Takaoka
- Unity Health Toronto, St Michael's Hospital, Toronto, Ontario, Canada
| | - Karen Colwill
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Vitaliy Matveev
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Feng Yun Yue
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Allison McGeer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Sharon Straus
- Unity Health Toronto, St Michael's Hospital, Toronto, Ontario, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Jane M. Heffernen
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, Toronto, Ontario, Canada
- Center for Disease Modelling, Mathematics and Statistics, York University, Toronto, Ontario, Canada
| | - Mario Ostrowski
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Unity Health Toronto, St Michael's Hospital, Toronto, Ontario, Canada
- Keenan Research Center for Biomedical Science, St Michael's Hospital Keenan, Toronto, Ontario, Canada
| |
Collapse
|
29
|
Fujitani M, Lu X, Shinnakasu R, Inoue T, Kidani Y, Seki NM, Ishida S, Mitsuki S, Ishihara T, Aoki M, Suzuki A, Takahashi K, Takayama M, Ota T, Iwata S, Shibata RY, Sonoyama T, Ariyasu M, Kitano A, Terooatea T, Kelly Villa J, Yamashita K, Yamasaki S, Kurosaki T, Omoto S. Longitudinal analysis of immune responses to SARS-CoV-2 recombinant vaccine S-268019-b in phase 1/2 prime-boost study. Front Immunol 2025; 16:1550279. [PMID: 40109335 PMCID: PMC11919840 DOI: 10.3389/fimmu.2025.1550279] [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: 12/23/2024] [Accepted: 02/13/2025] [Indexed: 03/22/2025] Open
Abstract
Background The durability of vaccine-induced immune memory to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is crucial for preventing infection, especially severe disease. Methods This follow-up report from a phase 1/2 study of S-268019-b (a recombinant spike protein vaccine) after homologous booster vaccination confirms its long-term safety, tolerability, and immunogenicity. Results Booster vaccination with S-268019-b resulted in an enhancement of serum neutralizing antibody (NAb) titers and a broad range of viral neutralization. Single-cell immune profiling revealed persistent and mature antigen-specific memory B cells and T follicular helper cells, with increased B-cell receptor diversity. The expansion of B- and T-cell repertoires and presence of cross-reactive NAbs targeting conserved epitopes within the receptor-binding domain following a booster accounted for the broad-spectrum neutralizing activity. Conclusion These findings highlight the potential of S-268019-b to provide broad and robust protection against a range of SARS-CoV-2 variants, addressing a critical challenge in the ongoing fight against coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Masaya Fujitani
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Xiuyuan Lu
- Laboratory of Molecular Immunology, World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Ryo Shinnakasu
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Takeshi Inoue
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Yujiro Kidani
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Naomi M. Seki
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Satoru Ishida
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Shungo Mitsuki
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | | | - Miwa Aoki
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Akio Suzuki
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Koji Takahashi
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Masahiro Takayama
- Pharmaceutical Technology Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Takeshi Ota
- Pharmaceutical Technology Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Satoshi Iwata
- Department of Microbiology, Tokyo Medical University, Tokyo, Japan
| | - Risa Yokokawa Shibata
- Drug Development and Regulatory Science Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Takuhiro Sonoyama
- Drug Development and Regulatory Science Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Mari Ariyasu
- Drug Development and Regulatory Science Division, Shionogi & Co., Ltd., Osaka, Japan
| | | | | | | | | | - Sho Yamasaki
- Laboratory of Molecular Immunology, World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan
| | - Shinya Omoto
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| |
Collapse
|
30
|
Cvijović I, Swift M, Quake SR. Long-term B cell memory emerges at uniform relative rates in the human immune response. Proc Natl Acad Sci U S A 2025; 122:e2406474122. [PMID: 40020190 PMCID: PMC11892634 DOI: 10.1073/pnas.2406474122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 01/13/2025] [Indexed: 03/12/2025] Open
Abstract
B cells generate pathogen-specific antibodies and play an essential role in providing adaptive protection against infection. Antibody genes are modified in evolutionary processes acting on the B cell populations within an individual. These populations proliferate, differentiate, and migrate to long-term niches in the body. However, the dynamics of these processes in the human immune system are primarily inferred from mouse studies. We addressed this gap by sequencing the antibody repertoire and transcriptomes from single B cells in four immune-rich tissues from six individuals. We find that B cells descended from the same pre-B cell ("lineages") often colocalize within the same tissue, with the bone marrow harboring the largest excess of lineages without representation in other tissues. Within lineages, cells with different levels of somatic hypermutation are uniformly distributed among tissues and functional states. This suggests that the relative probabilities of localization and differentiation outcomes change negligibly during affinity maturation, and quantitatively agrees with a simple dynamical model of B cell differentiation. While lineages strongly colocalize, we find individual B cells nevertheless appear to make independent differentiation decisions. Proliferative antibody-secreting cells, however, deviate from these global patterns. These cells are often clonally expanded, their clones appear universally distributed among all sampled organs, and form lineages with an excess of cells of the same type. Collectively, our findings show the limits of peripheral blood monitoring of the immune repertoire, and provide a probabilistic model of the dynamics of antibody memory formation in humans.
Collapse
Affiliation(s)
- Ivana Cvijović
- Department of Applied Physics, Stanford University, Stanford, CA94305
| | - Michael Swift
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA94305
| | - Stephen R. Quake
- Department of Applied Physics, Stanford University, Stanford, CA94305
- Department of Bioengineering, Stanford University, Stanford, CA94305
- The Chan Zuckerberg Initiative, Redwood City, CA94063
| |
Collapse
|
31
|
Górska A, Canziani LM, Rinaldi E, Pana ZD, Beale S, Bai F, Boxma-de Klerk BM, de Bruijn S, Donà D, Ekkelenkamp MB, Incardona F, Mallon P, Marchetti GC, Puhan M, Riva A, Simensen VC, Vaillant M, van der Zalm MM, van Kuijk SMJ, Wingerden SV, Judd A, Tacconelli E, Peñalvo JL. Learning from post-COVID-19 condition for epidemic preparedness: a variable catalogue for future post-acute infection syndromes. Clin Microbiol Infect 2025; 31:380-388. [PMID: 39662824 DOI: 10.1016/j.cmi.2024.12.001] [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: 09/12/2024] [Revised: 11/13/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024]
Abstract
SCOPE The emergence of post-COVID-19 condition (PCC) after SARS-CoV-2 infection underscores the critical need for preparedness in addressing future post-acute infection syndromes (PAIS), particularly those linked to epidemic outbreaks. The lack of standardized clinical and epidemiological data during the COVID-19 pandemic has significantly hindered timely diagnosis and effective treatment of PCC, highlighting the necessity of pre-emptively standardizing data collection in clinical studies to better define and manage future PAIS. In response, the Cohort Coordination Board, a consortium of European-funded COVID-19 research projects, has reviewed data from PCC studies conducted by its members. This paper leverages the Cohort Coordination Board's expertise to propose a standardized catalogue of variables, informed by the lessons learned during the pandemic, intended for immediate use in the design of future observational studies and clinical trials for emerging infections of epidemic potential. RECOMMENDATIONS The early implementation of standardized data collection, facilitated by the PAIS data catalogue, is essential for accelerating the identification and management of PAIS in future epidemics. This approach will enable more precise syndrome definitions, expedite diagnostic processes, and optimize treatment strategies, while also supporting long-term follow-up of affected individuals. The availability of harmonized data collection protocols will enhance preparedness across European and international cohort studies, and trials enabling a prompt and coordinated response, as well as more efficient resource allocation, in the event of emerging infections and associated PAIS.
Collapse
Affiliation(s)
- Anna Górska
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | | | - Eugenia Rinaldi
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Zoi D Pana
- Medical School, Basic and Clinical Studies Department, University of Nicosia, Nicosia, Cyprus
| | - Sarah Beale
- UCL Institute of Health Informatics, University College London, London, United Kingdom
| | - Francesca Bai
- Clinic of Infectious Diseases, San Paolo Hospital, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Bianca M Boxma-de Klerk
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Simeon de Bruijn
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Daniele Donà
- Department of Women's and Children's Health, Università Degli Studi di Padova, Padova, Italy
| | - Miquel B Ekkelenkamp
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Patrick Mallon
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
| | - Giulia C Marchetti
- Clinic of Infectious Diseases, San Paolo Hospital, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy; Department of Health Science, University of Milan, Milan, Italy
| | - Milo Puhan
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Agostino Riva
- Department of Biomedical and Clinical Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Victoria C Simensen
- Department of Vaccines and Immunisation, Division of Infectious Disease Control, Norwegian Institute of Public Health, Oslo, Norway
| | - Michel Vaillant
- Competence Centre for Methodology and Statistics, Department of Medical Informatics, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Marieke M van der Zalm
- Department of Paediatrics and Child Health, Stellenbosch University, Cape Town, South Africa
| | | | - Sophie van Wingerden
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Ali Judd
- MRC Clinical Trials Unit, University College London, London, United Kingdom; Fondazione Penta ETS, Padova, Italy
| | - Evelina Tacconelli
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - José L Peñalvo
- National Center for Epidemiology, Carlos III Health Institute (ISCIII), Madrid, Spain.
| |
Collapse
|
32
|
IJspeert H, Edwards ESJ, O'Hehir RE, Dalm VASH, van Zelm MC. Update on inborn errors of immunity. J Allergy Clin Immunol 2025; 155:740-751. [PMID: 39724969 DOI: 10.1016/j.jaci.2024.12.1075] [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: 11/15/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024]
Abstract
Ever since the first description of an inherited immunodeficiency in 1952 in a boy with gammaglobulin deficiency, new insights have progressed rapidly in disorders that are now referred to as inborn errors of immunity. In a field where fundamental molecular biology, genetics, immune signaling, and clinical care are tightly intertwined, 2022-24 saw a multitude of advances. Here we report a selection of research updates with a main focus on (1) diagnosis and screening, (2) new genetic defects, (3) susceptibility to severe coronavirus disease 2019 infection and impact of vaccination, and (4) treatment. Importantly, new pathogenic insights more rapidly affect treatment outcomes, either through an earlier and more precise diagnosis or through implementation of novel, personalized treatment. The field is growing rapidly, so awareness, communication, and collaboration are key to improving treatment outcomes.
Collapse
Affiliation(s)
- Hanna IJspeert
- Department of Clinical Genetics, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Emily S J Edwards
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Australia; Allergy, Asthma and Clinical Immunology, Alfred Health, Melbourne, Australia; Jeffrey Modell Center, Melbourne, Australia
| | - Robyn E O'Hehir
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Australia; Allergy, Asthma and Clinical Immunology, Alfred Health, Melbourne, Australia; Jeffrey Modell Center, Melbourne, Australia
| | - Virgil A S H Dalm
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands; Department of Internal Medicine, Division of Allergy & Clinical Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Menno C van Zelm
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands; Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Australia; Allergy, Asthma and Clinical Immunology, Alfred Health, Melbourne, Australia; Jeffrey Modell Center, Melbourne, Australia.
| |
Collapse
|
33
|
Das P, Varun CN, Ashwini M, Kesavan M, Ravi V, Desai A. Immunological memory in infected & exposure näive individuals one year post SARS-CoV-2 vaccination. Indian J Med Res 2025; 161:287-297. [PMID: 40347503 PMCID: PMC12066133 DOI: 10.25259/ijmr_923_2024] [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: 07/06/2024] [Accepted: 03/11/2025] [Indexed: 05/14/2025] Open
Abstract
Background & objectives COVID-19 has affected millions and wreaked havoc on global healthcare systems as well as a devastating impact on the economies of various nations. Vaccines are highly pivotal in promoting an appropriate immune response. Understanding the effectiveness and stability of the vaccines is essential as these may differ across populations. Hence, this study explored the durability and efficacy of the COVISHIELD (ChAdOx1 nCoV-A) vaccine among healthcare workers (HCWs). Methods In this study, 84 HCWs who received two doses of COVISHIELD and had no breakthrough infections or precautionary doses were assessed. Participants were categorised into four groups (A, B, C, and D) based on pre-immunisation antibody status and SARS-CoV-2 RT-PCR results. Group A had prior infection and IgG antibodies, group B had RT-PCR positivity without detectable antibodies, group C had IgG antibodies without RT-PCR testing, and group D was exposure-näive. Humoral immunity was assessed by measuring IgG antibodies to the spike protein, while cell-mediated immunity was evaluated through SARS-CoV-2-specific T-cell markers and immunophenotyping. Results Over one year, a significant decrease in anti-spike IgG levels was observed in all groups (P<0.0001). It was observed that memory B cells declined significantly among all the groups over a period of one year [group A (P<0.0001), group B (P=0.0080), group C (P=0.0158), and group D (P=0.0004)]; no significant decrease inactivated T-cells was observed over a year. The mean of anti-spike IgG levels in samples from exposed participants (group A, B, C) versus non-exposed (group D) was significantly higher (P<0.0001). Interpretation & conclusions These findings indicate the need for regular booster doses of vaccination due to the waning of immunity by the vaccine. We also demonstrate that hybrid immunity (a combination of immune response post-natural infection and vaccination) provides better protection than vaccination alone.
Collapse
Affiliation(s)
- Priti Das
- Department of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Chakrakodi N. Varun
- Department of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - M.A. Ashwini
- Department of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Muralidharan Kesavan
- Department of Psychiatry, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Vasanthapuram Ravi
- Department of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Anita Desai
- Department of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| |
Collapse
|
34
|
Raharinirina NA, Gubela N, Börnigen D, Smith MR, Oh DY, Budt M, Mache C, Schillings C, Fuchs S, Dürrwald R, Wolff T, Hölzer M, Paraskevopoulou S, von Kleist M. SARS-CoV-2 evolution on a dynamic immune landscape. Nature 2025; 639:196-204. [PMID: 39880955 PMCID: PMC11882442 DOI: 10.1038/s41586-024-08477-8] [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: 09/18/2023] [Accepted: 12/02/2024] [Indexed: 01/31/2025]
Abstract
Since the onset of the pandemic, many SARS-CoV-2 variants have emerged, exhibiting substantial evolution in the virus' spike protein1, the main target of neutralizing antibodies2. A plausible hypothesis proposes that the virus evolves to evade antibody-mediated neutralization (vaccine- or infection-induced) to maximize its ability to infect an immunologically experienced population1,3. Because viral infection induces neutralizing antibodies, viral evolution may thus navigate on a dynamic immune landscape that is shaped by local infection history. Here we developed a comprehensive mechanistic model, incorporating deep mutational scanning data4,5, antibody pharmacokinetics and regional genomic surveillance data, to predict the variant-specific relative number of susceptible individuals over time. We show that this quantity precisely matched historical variant dynamics, predicted future variant dynamics and explained global differences in variant dynamics. Our work strongly suggests that the ongoing pandemic continues to shape variant-specific population immunity, which determines a variant's ability to transmit, thus defining variant fitness. The model can be applied to any region by utilizing local genomic surveillance data, allows contextualizing risk assessment of variants and provides information for vaccine design.
Collapse
Affiliation(s)
- N Alexia Raharinirina
- Department of Mathematics & Computer Science, Freie Universität Berlin, Berlin, Germany
| | - Nils Gubela
- Department of Mathematics & Computer Science, Freie Universität Berlin, Berlin, Germany
- International Max-Planck Research School for Biology and Computation (IMPRS-BAC), Max-Planck Institute for Molecular Genetics, Berlin, Germany
| | | | | | - Djin-Ye Oh
- Department 1, Robert Koch Institute, Berlin, Germany
| | - Matthias Budt
- Department 1, Robert Koch Institute, Berlin, Germany
| | | | - Claudia Schillings
- Department of Mathematics & Computer Science, Freie Universität Berlin, Berlin, Germany
| | - Stephan Fuchs
- Department MFI, Robert Koch Institute, Berlin, Germany
| | - Ralf Dürrwald
- Department 1, Robert Koch Institute, Berlin, Germany
| | | | - Martin Hölzer
- Department MFI, Robert Koch Institute, Berlin, Germany
| | | | - Max von Kleist
- Department of Mathematics & Computer Science, Freie Universität Berlin, Berlin, Germany.
- Project Groups, Robert Koch Institute, Berlin, Germany.
| |
Collapse
|
35
|
Kovács Á, Hérincs Z, Papp K, Kaczmarek JZ, Larsen DN, Stage P, Bereczki L, Ujhelyi E, Pfeil T, Prechl J. In-depth immunochemical characterization of the serum antibody response using a dual-titration microspot assay. Front Immunol 2025; 16:1494624. [PMID: 40070838 PMCID: PMC11893856 DOI: 10.3389/fimmu.2025.1494624] [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: 09/11/2024] [Accepted: 02/03/2025] [Indexed: 03/14/2025] Open
Abstract
Antigen specific humoral immunity can be characterized by the analysis of serum antibodies. While serological assays for the measurement of specific antibody levels are available, these are not quantitative in the biochemical sense. Yet, understanding humoral immune responses quantitatively on the systemic level would need a universal, complete, quantitative, comparable measurement method of antigen specific serum antibodies of selected immunoglobulin classes. Here we describe a fluorescent, dual-titration immunoassay, which provides the biochemical parameters that are both necessary and sufficient to quantitatively characterize the humoral immune response. For validation of theory, we used recombinant receptor binding domain of SARS-CoV-2 as antigen on microspot arrays and varied the concentration of both the antigen and the serum antibodies from infected persons to obtain a measurement matrix of binding data. Both titration curves were simultaneously fitted using an algorithm based on the generalized logistic function and adapted for analyzing biochemical variables of binding. We obtained equilibrium affinity constants and concentrations for distinct antibody classes. These variables reflect the quality and the effective quantity of serum antibodies, respectively. The proposed fluorescent dual-titration microspot immunoassay can generate truly quantitative serological data that is suitable for immunological, medical and systems biological analysis.
Collapse
Affiliation(s)
- Ágnes Kovács
- Department of Biostatistics, University of Veterinary Medicine Budapest, Budapest, Hungary
- Department of Applied Analysis and Computational Mathematics, Eötvös Loránd University, Budapest, Hungary
| | | | | | | | - Daniel Nyberg Larsen
- Research and Development Department, Ovodan Biotech A/S, Odense, Denmark
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Pernille Stage
- Research and Development Department, Ovodan Biotech A/S, Odense, Denmark
| | | | | | - Tamás Pfeil
- Department of Applied Analysis and Computational Mathematics, Eötvös Loránd University, Budapest, Hungary
- HUN-REN–ELTE Numerical Analysis and Large Networks Research Group, Budapest, Hungary
| | | |
Collapse
|
36
|
Wang X, Yang X, Zhang X, Yan H, Jin J, Ma Z, Duan J, Zhang G, Huang T, Li Y, Wu H, Zhang T, Zhu A, Jin C, Song X, Su B. Dynamic SARS-CoV-2-specific B-cell and T-cell responses induced in people living with HIV after a full course of inactivated SARS-CoV-2 vaccine. Front Immunol 2025; 16:1554409. [PMID: 40070834 PMCID: PMC11893571 DOI: 10.3389/fimmu.2025.1554409] [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: 01/02/2025] [Accepted: 02/04/2025] [Indexed: 03/14/2025] Open
Abstract
Objective Both B-cell- and T-cell-mediated immunity are crucial for the effective clearance of viral infection, but little is known about the dynamic characteristics of SARS-CoV-2-specific B-cell and T-cell responses in people living with HIV (PLWH) after a full course of inactivated SARS-CoV-2 vaccination. Methods In this study, fifty people living with HIV (PLWH) and thirty healthy controls (HCs) were enrolled to assess B-cell and T-cell responses at the day before the vaccination (T0), two weeks after the first dose (T1), two months after the first dose (T2), the day of the third dose (T3), one month after the third dose (T4), three months after the third dose (T5) and 12 months (T6) after the third dose. Results SARS-CoV-2-specific B-cell and T-cell responses were induced in people living with HIV (PLWH), and these responses lasted at least one year after the third vaccine dose. However, the peak frequencies of Spike-specific B-cell and T-cell responses in PLWH were lower than those in HIV-negative controls. In addition, the expansion of activated B cells, memory B cells and plasma cells after primary vaccination was observed, but the percentages of these cells were decreased at T6 and were comparable to those at T0. Additionally, the percentages of activated T cells, exhausted T cells and SARS-CoV-2-specific T cells with enhanced functional activity were increased following the administration of inactivated SARS-CoV-2 vaccine. In addition, PLWH had lower percentages of plasma cells, RBD-specific B cells, circulating Tfh (cTfh) cells and CD38+ cTfh cells, and the percentages of the latter two types of cells were positively correlated with the titer of neutralizing antibodies, indicating these differences may account for the weaker immune responses induced in PLWH. Conclusion These data suggest that specific B-cell and T-cell responses could be sustained for at least one year after receiving the third vaccination. Our findings emphasize that the weak SARS-CoV-2-specific B-cell and T-cell responses induced in PLWH have implications for clinical decision-making and public health policy for PLWH with respect to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Xiuwen Wang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Center for General Practice Medicine, Department of Rheumatology and Immunology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaodong Yang
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xin Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Hongxia Yan
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Junyan Jin
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Zhenglai Ma
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Junyi Duan
- Tian Yuan Studio, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Guanghui Zhang
- Tian Yuan Studio, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Tao Huang
- Tian Yuan Studio, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yongzheng Li
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Hao Wu
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Tong Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Aiwei Zhu
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Cong Jin
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiangrong Song
- Department of Critical Care Medicine, Frontiers Science Center for Disease related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bin Su
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Central Laboratory, Beijing Youan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
37
|
Zaslavsky ME, Craig E, Michuda JK, Sehgal N, Ram-Mohan N, Lee JY, Nguyen KD, Hoh RA, Pham TD, Röltgen K, Lam B, Parsons ES, Macwana SR, DeJager W, Drapeau EM, Roskin KM, Cunningham-Rundles C, Moody MA, Haynes BF, Goldman JD, Heath JR, Chinthrajah RS, Nadeau KC, Pinsky BA, Blish CA, Hensley SE, Jensen K, Meyer E, Balboni I, Utz PJ, Merrill JT, Guthridge JM, James JA, Yang S, Tibshirani R, Kundaje A, Boyd SD. Disease diagnostics using machine learning of B cell and T cell receptor sequences. Science 2025; 387:eadp2407. [PMID: 39977494 PMCID: PMC12061481 DOI: 10.1126/science.adp2407] [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: 04/03/2024] [Accepted: 11/29/2024] [Indexed: 02/22/2025]
Abstract
Clinical diagnosis typically incorporates physical examination, patient history, various laboratory tests, and imaging studies but makes limited use of the human immune system's own record of antigen exposures encoded by receptors on B cells and T cells. We analyzed immune receptor datasets from 593 individuals to develop MAchine Learning for Immunological Diagnosis, an interpretive framework to screen for multiple illnesses simultaneously or precisely test for one condition. This approach detects specific infections, autoimmune disorders, vaccine responses, and disease severity differences. Human-interpretable features of the model recapitulate known immune responses to severe acute respiratory syndrome coronavirus 2, influenza, and human immunodeficiency virus, highlight antigen-specific receptors, and reveal distinct characteristics of systemic lupus erythematosus and type-1 diabetes autoreactivity. This analysis framework has broad potential for scientific and clinical interpretation of immune responses.
Collapse
MESH Headings
- Humans
- Autoimmune Diseases/diagnosis
- Autoimmune Diseases/immunology
- B-Lymphocytes/immunology
- COVID-19/diagnosis
- COVID-19/immunology
- Diabetes Mellitus, Type 1/diagnosis
- Diabetes Mellitus, Type 1/immunology
- HIV Infections/diagnosis
- HIV Infections/immunology
- Influenza, Human/diagnosis
- Influenza, Human/immunology
- Lupus Erythematosus, Systemic/diagnosis
- Lupus Erythematosus, Systemic/immunology
- Machine Learning
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- SARS-CoV-2/immunology
- Infections/diagnosis
- Infections/immunology
Collapse
Affiliation(s)
| | - Erin Craig
- Department of Biomedical Data Science, Stanford University; Stanford, CA, USA
| | - Jackson K. Michuda
- Department of Biomedical Data Science, Stanford University; Stanford, CA, USA
| | - Nidhi Sehgal
- Department of Genetics, Stanford University; Stanford, CA, USA
- Department of Pathology, Stanford University; Stanford, CA, USA
| | - Nikhil Ram-Mohan
- Department of Emergency Medicine, Stanford University; Stanford, CA, USA
| | - Ji-Yeun Lee
- Department of Pathology, Stanford University; Stanford, CA, USA
| | - Khoa D. Nguyen
- Department of Pathology, Stanford University; Stanford, CA, USA
| | - Ramona A. Hoh
- Department of Pathology, Stanford University; Stanford, CA, USA
| | - Tho D. Pham
- Department of Pathology, Stanford University; Stanford, CA, USA
- Stanford Blood Center; Stanford, CA, USA
| | - Katharina Röltgen
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute; Allschwil, Switzerland
- University of Basel; Basel, Switzerland
| | - Brandon Lam
- Department of Pathology, Stanford University; Stanford, CA, USA
| | - Ella S. Parsons
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University; Stanford, CA, USA
| | - Susan R. Macwana
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation; Oklahoma City, OK, USA
| | - Wade DeJager
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation; Oklahoma City, OK, USA
| | - Elizabeth M. Drapeau
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
| | - Krishna M. Roskin
- Department of Pediatrics, University of Cincinnati, College of Medicine; Cincinnati, OH, USA
- Divisions of Biomedical Informatics and Immunobiology, Cincinnati Children’s Hospital Medical Center; Cincinnati, OH, USA
| | | | - M. Anthony Moody
- Department of Pediatrics, Duke University; Durham, NC, USA
- Duke Human Vaccine Institute, Duke University; Durham, NC, USA
- Department of Immunology, Duke University; Durham, NC, USA
| | - Barton F. Haynes
- Duke Human Vaccine Institute, Duke University; Durham, NC, USA
- Department of Immunology, Duke University; Durham, NC, USA
- Department of Medicine, Duke University; Durham, NC, USA
| | - Jason D. Goldman
- Swedish Center for Research and Innovation, Swedish Medical Center; Seattle, WA, USA
- Division of Allergy and Infectious Diseases, University of Washington; Seattle, WA, USA
| | - James R. Heath
- Institute for Systems Biology; Seattle, WA, USA
- Department of Bioengineering, University of Washington; Seattle, WA, USA
| | - R. Sharon Chinthrajah
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University; Stanford, CA, USA
| | - Kari C. Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health; Boston, MA, USA
- Division of Allergy and Inflammation, Beth Israel Deaconess Medical Center; Boston, MA, USA
| | - Benjamin A. Pinsky
- Department of Pathology, Stanford University; Stanford, CA, USA
- Department of Medicine, Stanford University; Stanford, CA, USA
| | | | - Scott E. Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
| | - Kent Jensen
- Department of Medicine, Stanford University; Stanford, CA, USA
| | - Everett Meyer
- Department of Medicine, Stanford University; Stanford, CA, USA
| | - Imelda Balboni
- Department of Pediatrics, Stanford University; Stanford, CA, USA
| | - Paul J Utz
- Department of Medicine, Stanford University; Stanford, CA, USA
| | - Joan T. Merrill
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation; Oklahoma City, OK, USA
- Department of Medicine, Grossman School of Medicine, New York University; New York, NY, USA
- Lupus Foundation of America; Washington, DC, USA
| | - Joel M. Guthridge
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation; Oklahoma City, OK, USA
| | - Judith A. James
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation; Oklahoma City, OK, USA
| | - Samuel Yang
- Department of Emergency Medicine, Stanford University; Stanford, CA, USA
| | - Robert Tibshirani
- Department of Biomedical Data Science, Stanford University; Stanford, CA, USA
- Department of Statistics, Stanford University; Stanford, CA, USA
| | - Anshul Kundaje
- Department of Computer Science, Stanford University; Stanford, CA, USA
- Department of Genetics, Stanford University; Stanford, CA, USA
| | - Scott D. Boyd
- Department of Pathology, Stanford University; Stanford, CA, USA
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University; Stanford, CA, USA
| |
Collapse
|
38
|
Claireaux M, Elias G, Kerster G, Kuijper LH, Duurland MC, Paul AGA, Burger JA, Poniman M, Olijhoek W, de Jong N, de Jongh R, Wynberg E, van Willigen HDG, Prins M, De Bree GJ, de Jong MD, Kuijpers TW, Eftimov F, van der Schoot CE, Rispens T, Garcia-Vallejo JJ, ten Brinke A, van Gils MJ, van Ham SM. Deep profiling of B cells responding to various pathogens uncovers compartments in IgG memory B cell and antibody-secreting lineages. SCIENCE ADVANCES 2025; 11:eado1331. [PMID: 39970201 PMCID: PMC11837990 DOI: 10.1126/sciadv.ado1331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 01/16/2025] [Indexed: 02/21/2025]
Abstract
Improving our understanding of B cell transition to memory B cells (MBCs) and antibody-secreting cells (ASCs) is crucial for clinical monitoring and vaccine strategies. To explore these dynamics, we compared prepandemic antigen responses (influenza hemagglutinin, respiratory syncytial virus fusion glycoprotein, and tetanus toxoid) with recently encountered severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antigen responses in convalescent COVID-19 patients using spectral flow cytometry. Our analysis revealed the CD43+CD71+IgG+ activated B cell subset, highly enriched for SARS-CoV-2 specificities, as a juncture for ASC and MBC differentiation, with CD86+ phenotypically similar to ASCs and CD86- to IgG+ MBCs. Moreover, subpopulations within IgG+ MBCs were further identified based on CD73 and CD24 expression. Activated MBCs (CD73-/CD24lo) were predominantly SARS-CoV-2-specific, while resting MBCs (CD73+/CD24hi) recognized prepandemic antigens. A CD95- subcluster within resting MBCs accounted for over 40% of prepandemic-specific cells, indicating long-lasting memory. These findings advance our understanding of IgG+ MBC and ASC development stages, shedding light on the decision-making process guiding their differentiation.
Collapse
Affiliation(s)
- Mathieu Claireaux
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
| | - George Elias
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
| | - Gius Kerster
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
| | - Lisan H. Kuijper
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
| | - Mariël C. Duurland
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
| | | | - Judith A. Burger
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
| | - Meliawati Poniman
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
| | - Wouter Olijhoek
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
| | - Nina de Jong
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
| | - Rivka de Jongh
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
| | - Elke Wynberg
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Infectious Diseases, Public Health Service of Amsterdam, GGD, Amsterdam, Netherlands
| | - Hugo D. G. van Willigen
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
| | - Maria Prins
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Infectious Diseases, Public Health Service of Amsterdam, GGD, Amsterdam, Netherlands
- Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Godelieve J. De Bree
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Menno D. de Jong
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
| | - Taco W. Kuijpers
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Pediatric Immunology, Rheumatology and Infectious Disease, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Filip Eftimov
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC, location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - C. Ellen van der Schoot
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
| | - Theo Rispens
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
| | - Juan J. Garcia-Vallejo
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Molecular Cell Biology & Immunology, Amsterdam University Medical Center (VUmc location), Amsterdam, Netherlands
- Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Anja ten Brinke
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
| | - Marit J. van Gils
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
| | - S. Marieke van Ham
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
39
|
Russo C, Otero A, Uranga M, Seery V, Raiden S, Algieri S, De Carli N, Borda M, Albistur MF, Heinitz L, Marcó del Pont M, Pardini M, Budano G, Alvarez L, Simaz N, Merhar C, Quintana MC, Garbini C, Portela LA, Pereira MS, Ferrero F, Geffner J, Arruvito L. Immunological memory to COVID-19 vaccines in immunocompromised and immunocompetent children. Front Cell Infect Microbiol 2025; 15:1527573. [PMID: 40034392 PMCID: PMC11873107 DOI: 10.3389/fcimb.2025.1527573] [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: 11/13/2024] [Accepted: 01/21/2025] [Indexed: 03/05/2025] Open
Abstract
Background Most children in Argentina received only the initial COVID-19 vaccine series, with presumed hybrid immunity after multiple Omicron waves. However, the durability of immune memory, particularly in immunocompromised (IC) children, remains poorly studied. Methods A cohort of IC (n=45) and healthy children (HC, n=79) was assessed between 13 to 17 months after receiving two or three doses of BBIBP-CorV and/or BNT162b2. Plasma anti-spike IgG, neutralizing activity and antigen-specific CD4+ and CD8+ T cells against Wuhan and Omicron BA.5 variants were assessed. Results Most children remained seropositive after two vaccine doses, but compared with HC, IC exhibited lower neutralizing titers against both Wuhan and Omicron BA.5, particularly those vaccinated with BBIBP-CorV. Even after three vaccine doses, IC showed weaker neutralizing antibody response, CD8+ T cell responses and lower IFN-γ production compared with HC. Integrated analysis of neutralizing antibodies, memory CD4+, and CD8+ T cells revealed a weak immune memory among IC with an important compromise in memory CD8+ T cell responses. Conclusions Immunity can last up to 17 months, but reduced effectiveness against new variants highlights the need for updated COVID-19 vaccines, especially for IC children. Additional efforts are essential to enhance vaccination coverage and protect this vulnerable population.
Collapse
Affiliation(s)
- Constanza Russo
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Facultad de Medicina, UBA-CONICET, Buenos Aires, Argentina
| | - Adrián Otero
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Facultad de Medicina, UBA-CONICET, Buenos Aires, Argentina
| | - Macarena Uranga
- Departamento de Medicina, Hospital Universitario Austral, Buenos Aires, Argentina
| | - Vanesa Seery
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Facultad de Medicina, UBA-CONICET, Buenos Aires, Argentina
| | - Silvina Raiden
- Departamento de Medicina, Hospital General de Niños Pedro de Elizalde, Buenos Aires, Argentina
| | - Silvia Algieri
- Servicio de Pediatria, Hospital Nacional Profesor Alejandro Posadas, Buenos Aires, Argentina
| | - Norberto De Carli
- Servicio de Pediatria, Clínica del Niño de Quilmes, Buenos Aires, Argentina
| | - Mauricio Borda
- Servicio de Pediatria, Hospital Pediátrico Juan Pablo II, Corrientes, Argentina
| | - María F. Albistur
- Departamento de Medicina, Hospital Universitario Austral, Buenos Aires, Argentina
| | - Lourdes Heinitz
- Departamento de Medicina, Hospital Universitario Austral, Buenos Aires, Argentina
| | - María Marcó del Pont
- Departamento de Medicina, Hospital Universitario Austral, Buenos Aires, Argentina
| | - Martina Pardini
- Departamento de Medicina, Hospital Universitario Austral, Buenos Aires, Argentina
| | - Guillermina Budano
- Departamento de Medicina, Hospital Universitario Austral, Buenos Aires, Argentina
| | - Laura Alvarez
- Departamento de Medicina, Hospital Universitario Austral, Buenos Aires, Argentina
| | - Nancy Simaz
- Servicio de Pediatria, Hospital Nacional Profesor Alejandro Posadas, Buenos Aires, Argentina
| | - Claudia Merhar
- Servicio de Pediatria, Hospital Nacional Profesor Alejandro Posadas, Buenos Aires, Argentina
| | - María C. Quintana
- Servicio de Pediatria, Hospital Nacional Profesor Alejandro Posadas, Buenos Aires, Argentina
| | - Cecilia Garbini
- Servicio de Pediatria, Hospital Nacional Profesor Alejandro Posadas, Buenos Aires, Argentina
| | - Luisa Aedo Portela
- Servicio de Pediatria, Clínica del Niño de Quilmes, Buenos Aires, Argentina
| | | | - Fernando Ferrero
- Departamento de Medicina, Hospital General de Niños Pedro de Elizalde, Buenos Aires, Argentina
| | - Jorge Geffner
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Facultad de Medicina, UBA-CONICET, Buenos Aires, Argentina
| | - Lourdes Arruvito
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Facultad de Medicina, UBA-CONICET, Buenos Aires, Argentina
| |
Collapse
|
40
|
Güzel I, Öztürk G, Appak Ö, Çağlayan D, Süner AF, Irmak Ç, Türe N, Işik E, Çelik M, Ergör G, Ergör A, Demiral Y, Alp Çavuş S, Kilic B, Sayiner A. Neutralizing and binding antibody dynamics following primary and booster COVID-19 vaccination among healthcare workers. BMC Infect Dis 2025; 25:218. [PMID: 39953414 PMCID: PMC11827177 DOI: 10.1186/s12879-025-10621-2] [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: 09/30/2024] [Accepted: 02/07/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Vaccine-induced neutralizing antibodies (NAbs) are key for COVID-19 protective-immunity. As the efficacy of SARS-CoV-2 vaccines declines over time and variants of the virus continue to emerge, the need for booster doses of vaccine remains on the agenda. The aim of this study was to assess NAbs dynamics and its correlation with anti-RBD IgG levels during the nine-month follow-up period after primary-CoronaVac vaccination and booster vaccinations to evaluate vaccination strategies. METHODS This prospective longitudinal observational study followed 226 healthcare workers who received primary (two doses CoronaVac) and booster (CoronaVac or BNT162b2) immunization. Serum samples were collected at four different time points, two after primary vaccination and two after booster. Anti-RBD IgG antibody levels were assessed with the SARS CoV-2 IgG-II-QUANT kit (Abbott, USA) and neutralizing antibody levels were determined with the ACE2-RBD-Neutralization-Assay (Dia-Pro, Italy) using a surrogate virus neutralization method. Factors affecting antibody response were analyzed. Statistical analysis was performed with IBM-SPSS-22.0. RESULTS One month after the second dose of CoronaVac, 79.2% of participants had NAb, but this had decreased to 49.7% by the fourth month and was influenced by smoking, BMI and chronic diseases. Boosters, regardless of type, significantly raised NAb levels. Heterologous vaccination yielded higher NAb and anti-RBD IgG responses. Both single or double-BNT162b2 boosters resulted in similar NAb responses. There was a strong correlation between anti-RBD IgG and NAb levels following CoronaVac vaccination, leading to the identification of predictive IgG threshold for the presence of NAb. The type of booster influenced the correlation strength and threshold-value. CONCLUSIONS NAbs levels decreased rapidly after primary CoronaVac vaccination. Boosters significantly increased levels while the heterologous vaccine combination induced a greater response. Anti-RBD IgG levels were able to predict the NAb response, however the correlation varied by the vaccine type, NAb response strength and the time since vaccination.
Collapse
Affiliation(s)
- Irmak Güzel
- Department of Medical Microbiology, Turkish Republic Ministry of Health, Nusaybin State Hospital, Nusaybin, Turkey
| | - Gamze Öztürk
- Department of Medical Microbiology, Faculty of Medicine, Dokuz Eylül University, Izmir, Turkey
| | - Özgür Appak
- Department of Medical Microbiology, Faculty of Medicine, Dokuz Eylül University, Izmir, Turkey
| | - Derya Çağlayan
- Department of Public Health, Division of Epidemiology, Turkish Republic Ministry of Health, Diyarbakır Provincial Health Directorate, Diyarbakır, Turkey
| | - Ahmet F Süner
- Çaycuma District Health Directorate, Zonguldak, Turkey
| | - Çağlar Irmak
- Infectious Diseases and Clinical Microbiology Unit, Hakkari Yüksekova State Hospital, Hakkari, Turkey
| | - Neslişah Türe
- Department of Public Health, Faculty of Medicine, Dokuz Eylül University, Izmir, Turkey
| | - Elif Işik
- Department of Public Health, Faculty of Medicine, Dokuz Eylül University, Izmir, Turkey
| | - Muammer Çelik
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Dokuz Eylül University, Izmir, Turkey
| | - Gül Ergör
- Department of Public Health, Division of Epidemiology, Faculty of Medicine, Dokuz Eylül University, Izmir, Turkey
| | - Alp Ergör
- Department of Public Health, Faculty of Medicine, Dokuz Eylül University, Izmir, Turkey
| | - Yücel Demiral
- Department of Public Health, Faculty of Medicine, Dokuz Eylül University, Izmir, Turkey
| | - Sema Alp Çavuş
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Dokuz Eylül University, Izmir, Turkey
| | - Bulent Kilic
- Department of Public Health, Faculty of Medicine, Dokuz Eylül University, Izmir, Turkey
| | - Arzu Sayiner
- Department of Medical Microbiology, Faculty of Medicine, Dokuz Eylül University, Izmir, Turkey.
| |
Collapse
|
41
|
Naiditch H, Betts MR, Larman HB, Levi M, Rosenberg AZ. Immunologic and inflammatory consequences of SARS-CoV-2 infection and its implications in renal disease. Front Immunol 2025; 15:1376654. [PMID: 40012912 PMCID: PMC11861071 DOI: 10.3389/fimmu.2024.1376654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 12/23/2024] [Indexed: 02/28/2025] Open
Abstract
The emergence of the COVID-19 pandemic made it critical to understand the immune and inflammatory responses to the SARS-CoV-2 virus. It became increasingly recognized that the immune response was a key mediator of illness severity and that its mechanisms needed to be better understood. Early infection of both tissue and immune cells, such as macrophages, leading to pyroptosis-mediated inflammasome production in an organ system critical for systemic oxygenation likely plays a central role in the morbidity wrought by SARS-CoV-2. Delayed transcription of Type I and Type III interferons by SARS-CoV-2 may lead to early disinhibition of viral replication. Cytokines such as interleukin-1 (IL-1), IL-6, IL-12, and tumor necrosis factor α (TNFα), some of which may be produced through mechanisms involving nuclear factor kappa B (NF-κB), likely contribute to the hyperinflammatory state in patients with severe COVID-19. Lymphopenia, more apparent among natural killer (NK) cells, CD8+ T-cells, and B-cells, can contribute to disease severity and may reflect direct cytopathic effects of SARS-CoV-2 or end-organ sequestration. Direct infection and immune activation of endothelial cells by SARS-CoV-2 may be a critical mechanism through which end-organ systems are impacted. In this context, endovascular neutrophil extracellular trap (NET) formation and microthrombi development can be seen in the lungs and other critical organs throughout the body, such as the heart, gut, and brain. The kidney may be among the most impacted extrapulmonary organ by SARS-CoV-2 infection owing to a high concentration of ACE2 and exposure to systemic SARS-CoV-2. In the kidney, acute tubular injury, early myofibroblast activation, and collapsing glomerulopathy in select populations likely account for COVID-19-related AKI and CKD development. The development of COVID-19-associated nephropathy (COVAN), in particular, may be mediated through IL-6 and signal transducer and activator of transcription 3 (STAT3) signaling, suggesting a direct connection between the COVID-19-related immune response and the development of chronic disease. Chronic manifestations of COVID-19 also include systemic conditions like Multisystem Inflammatory Syndrome in Children (MIS-C) and Adults (MIS-A) and post-acute sequelae of COVID-19 (PASC), which may reflect a spectrum of clinical presentations of persistent immune dysregulation. The lessons learned and those undergoing continued study likely have broad implications for understanding viral infections' immunologic and inflammatory consequences beyond coronaviruses.
Collapse
Affiliation(s)
- Hiam Naiditch
- Department of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael R. Betts
- Department of Microbiology and Institute of Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - H. Benjamin Larman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Moshe Levi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
| | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
42
|
Trimbake D, Singh D, K. YG, Babar P, S. VD, Tripathy AS. Durability of Functional SARS-CoV-2-Specific Immunological Memory and T Cell Response up to 8-9 Months Postrecovery From COVID-19. J Immunol Res 2025; 2025:9743866. [PMID: 39963186 PMCID: PMC11832264 DOI: 10.1155/jimr/9743866] [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: 06/03/2024] [Accepted: 12/19/2024] [Indexed: 02/20/2025] Open
Abstract
Research on long-term follow-up in individuals who have recovered from coronavirus disease-19 (COVID-19) would yield insights regarding their immunity status and identify those who need booster vaccinations. This study evaluated the longevity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific cellular and humoral memory responses, as well as T cell effector functionalities, at 1-2 months (n = 40), 8-9 months (n = 40), and 12 months/1 year (n = 27) following recovery from SARS-CoV-2 infection. CTL response by enzyme-linked immunospot (ELISPOT); levels of cytokine by Bio-Plex, natural killer (NK), CD4+ helper, and CD8+ cytotoxic T cell functionalities using flow cytometry; anti-SARS-CoV-2 IgG by ELISA; and levels of neutralizing antibodies (NAbs) by surrogate virus NAb assay were assessed. The levels of SARS-CoV-2-specific IgG and NAb at 1-2 and 8-9 months postrecovery were hand in hand and appeared declining. SARS-CoV-2-specific B, memory B and plasma cells, and T cells sustained up to 8-9 months. Increased expression of CD107a/IFN-γ by NK cells and cytotoxic T cells at 8-9 months could be indicative of SARS-CoV-2-specific effector functions. Recovered individuals with positive and negative IgG antibody status displayed T cell response up to 1 year and 8-9 months, respectively, emphasizing the durabilty of effector immunity up to 8-9 months regardless of IgG antibody status. Overall, the recovered individuals exhibited robust immunological memory, sustained T cell response with effector functionality against SARS-CoV-2 that persists for at least 8-9 months.
Collapse
Affiliation(s)
- Diptee Trimbake
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| | - Dharmendra Singh
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| | - Yogesh Gurav K.
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| | - Prasad Babar
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| | - Varsha Dange S.
- Department of Medicine, Pimpri Chinchwad Municipal Corporation, Pimpri, Pune 411018, Maharashtra, India
| | - Anuradha S. Tripathy
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| |
Collapse
|
43
|
Ledford H. Four lessons COVID taught us about the immune system. Nature 2025; 638:19-20. [PMID: 39870893 DOI: 10.1038/d41586-025-00128-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
|
44
|
Mendoza-Ramírez NJ, García-Cordero J, Hernández-Galicia G, Moreno-Licona NJ, Hernandez J, Cabello-Gutierrez C, Zúñiga-Ramos JA, Morales-Rios E, Pérez-Tapia SM, Ortiz-Navarrete V, Espinosa-Cantellano M, Fernández-Benavides DA, Cedillo-Barrón L. Vaccination with Plasmids Encoding the Fusion Proteins D-S1, D-S1N and O-SN from SARS-CoV-2 Induces an Effective Humoral and Cellular Immune Response in Mice. Vaccines (Basel) 2025; 13:134. [PMID: 40006682 PMCID: PMC11860763 DOI: 10.3390/vaccines13020134] [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: 12/10/2024] [Revised: 01/08/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Next-generation vaccines against coronavirus disease 2019 (COVID-19) focus on inducing a long-lasting immune response against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and its emerging variants. To achieve this, antigens other than spike proteins have been proposed, and different platforms have been evaluated. Nucleic acid-based vaccines are fundamental for this process. Preclinical data have shown that the SARS-CoV-2 nucleocapsid protein induces a protective cellular immune response, and when combined with the spike protein, the resulting humoral and cellular immune responses are effective against some SARS-CoV-2 variants. METHODS We designed a DNA vaccine against the spike and nucleocapsid proteins of SARS-CoV-2 to generate fusion proteins based on the Delta and Omicron B.5 strains. The most immunogenic regions of the spike and nucleocapsid proteins of the Delta and Omicron B strains were selected using bioinformatics. The nucleotide sequences were cloned into pcDNA3.1, and named pcDNA3.1/D-S1, pcDNA3.1/D-S1N, and pcDNA3.1/O-SN. The immunogenicity of the generated fusion proteins was evaluated by analyzing the humoral and cellular responses elicited after the immunization of BALB/c mice. RESULTS DNA immunization induced antibody production, neutralization activity, and IFN-γ production. The inclusion of the nucleocapsid regions in the plasmid greatly enhanced the immune response. Moreover, cross-reactions with the variants of interest were confirmed. CONCLUSIONS Plasmids-encoding fusion proteins combining the most immunogenic regions of the spike and nucleocapsid proteins present a promising strategy for designing new and effective vaccines against SARS-CoV-2.
Collapse
Affiliation(s)
- Noe Juvenal Mendoza-Ramírez
- Departamento de Biomedicina Molecular CINVESTAV, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico; (N.J.M.-R.); (J.G.-C.); (G.H.-G.)
| | - Julio García-Cordero
- Departamento de Biomedicina Molecular CINVESTAV, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico; (N.J.M.-R.); (J.G.-C.); (G.H.-G.)
| | - Gabriela Hernández-Galicia
- Departamento de Biomedicina Molecular CINVESTAV, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico; (N.J.M.-R.); (J.G.-C.); (G.H.-G.)
| | - Nicole Justine Moreno-Licona
- Departamento de Bioquímica Cinvestav, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico; (N.J.M.-L.); (E.M.-R.)
| | - Jesus Hernandez
- Laboratorio de Inmunología, Centro de Investigación en Alimentación y Desarrollo A. C (CIAD) Carretera a la Victoria km 0.6, Hermosillo Sonora 83304, Mexico
| | - Carlos Cabello-Gutierrez
- Departamento de Investigación en Virología y Micología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Calzada de Tlalpan 4502, Belisario Domínguez, Tlalpan 14080, Mexico; (C.C.-G.); (J.A.Z.-R.)
| | - Joaquín Alejandro Zúñiga-Ramos
- Departamento de Investigación en Virología y Micología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Calzada de Tlalpan 4502, Belisario Domínguez, Tlalpan 14080, Mexico; (C.C.-G.); (J.A.Z.-R.)
- Escuela de Medicina y Ciencias de la Salud, Tecnológico de Monterrey, Monterrey 64710, Mexico
| | - Edgar Morales-Rios
- Departamento de Bioquímica Cinvestav, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico; (N.J.M.-L.); (E.M.-R.)
| | - Sonia Mayra Pérez-Tapia
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City 11340, Mexico;
| | - Vianney Ortiz-Navarrete
- Departamento de Biomedicina Molecular CINVESTAV, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico; (N.J.M.-R.); (J.G.-C.); (G.H.-G.)
| | - Martha Espinosa-Cantellano
- Departamento de Infectómica y Patogénesis Molecular, CINVESTAV, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico;
| | - David Andrés Fernández-Benavides
- Centro de Ingeniería y Desarrollo Industrial (CIDESI), Av. Playa Pie de la Cuesta No. 702, Desarrollo San Pablo, Querétaro 76125, Mexico;
| | - Leticia Cedillo-Barrón
- Departamento de Biomedicina Molecular CINVESTAV, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico; (N.J.M.-R.); (J.G.-C.); (G.H.-G.)
| |
Collapse
|
45
|
Hromić-Jahjefendić A, Aljabali AAA. Analysis of the immune response in COVID-19. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:31-71. [PMID: 40246347 DOI: 10.1016/bs.pmbts.2024.10.004] [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: 04/19/2025]
Abstract
The COVID-19 pandemic, instigated by the novel coronavirus SARS-CoV-2, has emerged as a significant global health challenge, demanding a profound grasp of the immune response. The innate immune system, a multifaceted network encompassing pattern recognition receptors (PRRs) and effector cells, assumes a pivotal function in detecting and countering this viral assailant. Toll-like receptors (TLRs), situated on immune cell surfaces and within endosomes, play a central role in recognizing SARS-CoV-2. TLR-2 and TLR-4 discern specific viral constituents, such as the spike (S) protein, setting off inflammatory signaling cascades and catalyzing the generation of type I interferons. Intracellular PRRs, including the RIG-I-like receptors (RLRs), RIG-I and MDA5, detect viral RNA within the cytoplasm of infected cells, provoking antiviral responses by initiating the synthesis of type I interferons. The equilibrium between interferons and pro-inflammatory cytokines dictates the outcomes of the disease. Interferons play an indispensable role in governing viral replication, while unregulated cytokine production can result in tissue harm and inflammation. This intricate dynamic underpins therapeutic strategies aimed at regulating immune responses in individuals grappling with COVID-19. Natural killer (NK) cells, with their capacity to recognize infected cells through the "missing self" phenomenon and activating receptors, make significant contributions to the defense against SARS-CoV-2. NK cells play a pivotal role in eliminating infected cells and boosting immune responses through antibody-dependent cell-mediated cytotoxicity (ADCC). In conclusion, comprehending the interplay among PRRs, interferons, and NK cells within innate immunity is paramount for discerning and combatting SARS-CoV-2. This comprehension illuminates therapeutic interventions and vaccine development, casting light on our endeavors to confront this worldwide health crisis.
Collapse
Affiliation(s)
- Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina.
| | - Alaa A A Aljabali
- Faculty of Pharmacy, Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid, Jordan
| |
Collapse
|
46
|
Hoffmann DJ, Meyer PWA, Worsley CM, van der Mescht MA, Visser A, Pillay TS. SARS-CoV-2 seropositivity amongst healthcare workers in South Africa during the Omicron wave: natural infection versus vaccination. J Clin Pathol 2025:jcp-2024-209722. [PMID: 39824540 DOI: 10.1136/jcp-2024-209722] [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: 06/21/2024] [Accepted: 12/09/2024] [Indexed: 01/20/2025]
Abstract
AIMS Concerns over population-level immunity have been heightened with each successive wave of COVID-19, prompting questions about whether it is primarily derived from vaccination efforts or from previous natural infections with the virus. We wished to determine the seroprevalence of SARS-CoV-2 antibodies among healthcare workers (HCWs) in Pretoria (Tshwane), South Africa, and to establish whether they were derived from vaccination or natural infection. METHODS Serum samples were collected from HCWs during the fourth wave of COVID-19 between 1 December 2021 and 13 March 2022. The samples were tested using the Abbott SARS-CoV-2 Spike IgG (S-IgG), IgM (S-IgM) and the SARS-CoV-2 Nucleocapsid IgG (NC-IgG) kits. RESULTS Of the 221 participants, 76% (n=168) were women and 24% (n=53) were men. A total of 96.4% (n=213) of the participants were vaccinated. Natural infection-derived antibodies were detected in 23% (n=51) of participants, and vaccine-derived antibodies in 74% (n=164) of the HCWs. CONCLUSIONS Even after three waves of COVID-19, HCWs derived most of their detectable antibodies from vaccination. Vaccination remains an essential tool to protect HCWs and patients from SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Daniel J Hoffmann
- Department of Clinical Pathology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- National Health Laboratory Service, Pretoria, South Africa
| | - Pieter W A Meyer
- National Health Laboratory Service, Pretoria, South Africa
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Catherine M Worsley
- National Health Laboratory Service, Pretoria, South Africa
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Mieke A van der Mescht
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - A Visser
- Department of Clinical Pathology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- National Health Laboratory Service, Pretoria, South Africa
| | - Tahir S Pillay
- Department of Clinical Pathology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- National Health Laboratory Service, Pretoria, South Africa
- Department of Chemical Pathology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
47
|
Oliva-Ariza G, Criado I, Fuentes-Herrero B, Carbonell C, Sánchez-Gallego JI, López-Bernús A, Gutiérrez ML, Rolo-Ramírez A, Bernal-Ribes M, Almenara-Morales Y, Lecrevisse Q, van Dongen JJM, Marcos M, Almeida J, Orfao A. Early Immune Cell and Antibody Kinetics Following SARS-CoV-2 Vaccination in Healthy Adults and Low-Count Monoclonal B-Cell Lymphocytosis. Int J Mol Sci 2025; 26:681. [PMID: 39859394 PMCID: PMC11765611 DOI: 10.3390/ijms26020681] [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: 12/09/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
The early immune kinetics after SARS-CoV-2 vaccination remain poorly understood, particularly among individuals with low-count monoclonal B-cell lymphocytosis (MBLlo). We investigated the cellular and humoral kinetics in the blood of 50 non-MBL healthy donors (HD) vs. 16 MBLlo subjects after SARS-CoV-2 vaccination, who were subclassified according to their history of previous exposure to SARS-CoV-2 into SARS-CoV-2 naïve and previously infected subjects. Overall, we found decreased neutrophil and lymphocyte counts at day +4 following each dose in non-MBL HD, together with an earlier and higher increase in plasma cell (PC) counts and SARS-CoV-2-specific antibody levels after the first vaccine in previously infected non-MBL HD. MBLlo subjects showed a similar profile, except for lower B-cell and higher PC counts after vaccination, and a trend towards a higher (but delayed) antibody response. In summary, we found different cell-kinetic profiles following vaccination in SARS-CoV-2 naïve vs. previously infected non-MBL HD (earlier PC and antibody responses in the latter group); additionally, MBLlo subjects had significantly lower B-cell and higher PC counts after vaccination, and a delayed SARS-CoV-2-specific antibody response.
Collapse
Affiliation(s)
- Guillermo Oliva-Ariza
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Ignacio Criado
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Blanca Fuentes-Herrero
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Cristina Carbonell
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Department of Internal Medicine, University Hospital of Salamanca, 37007 Salamanca, Spain; (A.R.-R.); (Y.A.-M.)
- Department of Infectious Diseases, University Hospital of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), 37007 Salamanca, Spain
| | - José Ignacio Sánchez-Gallego
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Amparo López-Bernús
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Department of Internal Medicine, University Hospital of Salamanca, 37007 Salamanca, Spain; (A.R.-R.); (Y.A.-M.)
- Department of Infectious Diseases, University Hospital of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), 37007 Salamanca, Spain
| | - María Laura Gutiérrez
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alejandro Rolo-Ramírez
- Department of Internal Medicine, University Hospital of Salamanca, 37007 Salamanca, Spain; (A.R.-R.); (Y.A.-M.)
| | - Marta Bernal-Ribes
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Yolimar Almenara-Morales
- Department of Internal Medicine, University Hospital of Salamanca, 37007 Salamanca, Spain; (A.R.-R.); (Y.A.-M.)
| | - Quentin Lecrevisse
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jacques J. M. van Dongen
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
| | - Miguel Marcos
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Department of Internal Medicine, University Hospital of Salamanca, 37007 Salamanca, Spain; (A.R.-R.); (Y.A.-M.)
| | - Julia Almeida
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alberto Orfao
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
48
|
Thiem VD, Anh DD, Ha VH, Van Thom N, Thang TC, Mateus J, Carreño JM, Raghunandan R, Huong NM, Mercer LD, Flores J, Escarrega EA, Raskin A, Thai DH, Van Be L, Sette A, Innis BL, Krammer F, Weiskopf D. Safety and immunogenicity of an inactivated recombinant Newcastle disease virus vaccine expressing SARS-CoV-2 spike: A randomised, comparator-controlled, phase 2 trial. Vaccine 2025; 44:126542. [PMID: 39615342 PMCID: PMC11672239 DOI: 10.1016/j.vaccine.2024.126542] [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: 05/20/2024] [Revised: 11/11/2024] [Accepted: 11/16/2024] [Indexed: 12/20/2024]
Abstract
Production of affordable coronavirus disease 2019 (COVID-19) vaccines in low- and lower-middle-income countries is needed. NDV-HXP-S is an inactivated egg-based recombinant Newcastle disease virus vaccine expressing the spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). A public sector manufacturer in Vietnam assessed the immunogenicity of NDV-HXP-S (COVIVAC) relative to an authorized vaccine. This phase 2 stage of a randomised, observer-blind, controlled, phase 1/2 trial was conducted at three community health centers in Thai Binh Province, Vietnam. Healthy males and non-pregnant females, 18 years of age and older, were eligible. Participants were randomised by age (18-59, ≥60 years) to receive one of three treatments by intramuscular injection twice, 28 days apart: COVIVAC at 3 μg or 6 μg, or AstraZeneca COVID-19 vaccine VAXZEVRIA™. Participants and personnel assessing outcomes were masked to treatment. The vaccine dose was selected based on Phase 1 results. A 6 μg dose was chosen to explore the immunogenicity gain over the 3-μg dose. The study's aim is to evaluate the safety and immunogenicity of COVIVAC at two dose levels compared to VAXZEVRIA, the most commonly used COVID-19 vaccine in Vietnam. The main outcome was the induction of 50% neutralising antibody titers against vaccine-homologous pseudotyped virus 14 days (day 43) and 6 months (day 197) after the second vaccination by age group. The primary immunogenicity and safety analyses included all participants who received one dose of the vaccine. ClinicalTrials.govNCT05940194. During August 10-23, 2021, 737 individuals were screened, and 374 were randomised (124-125 per group); all subjects received vaccine dose one and all but three received doses two four weeks later. Subjects 18-59 years of age achieved the following geometric mean titers of PNA 14 days after vaccine dose two: 153⋅28 (95 % CI 124·2-189⋅15) for COVIVAC 3 μg, 176⋅2 (95 % CI 141⋅45-220.27) for COVIVAC 6 μg, and 99⋅92(95 % CI 80.80-123⋅56) for VAXZEVRIA. Subjects ≥60 years of age also achieved potent geometric mean titers of PNA at the same timepoint: 183⋅57 (95 % CI 133.4-252⋅61) for COVIVAC 3 μg, 257⋅87 (95 % CI 181⋅6-367⋅18) for COVIVAC 6 μg, and 79⋅49(95 % CI 55⋅68-113⋅4) for VAXZEVRIA. On day 43, the geometric mean fold rise of 50 % neutralising antibody titers for subjects age 18-59 years was 31·20 (COVIVAC 3 μg N = 82, 95 % CI 25·14-38·74), 35·80 (COVIVAC 6 μg; N = 83, 95 % CI 29·03-44·15), 18·85 (VAXZEVRIA; N = 82, 95 % CI 15·10-23·54), and for subjects age ≥ 60 years was 37·27 (COVIVAC 3 μg; N = 42, 95 % CI 27·43-50·63), 50·10 (COVIVAC 6 μg; N = 40, 95 % CI 35·46-70·76), 16·11 (VAXZEVRIA; N = 40, 95 % CI 11·73-22·13). Among subjects seronegative for anti-S IgG at baseline, the day 43 geometric mean titer ratio of neutralising antibody (COVIVC 6 μg/VAXZEVRIA) was 1·77 (95 % CI 1·30-2·40) for subjects age 18-59 years and 3·24 (95 % CI 1·98-5·32) for subjects age ≥ 60 years. On day 197, the age-specific ratios were 1·11 (95 % CI 0·51-2·43) and 2·32 (0·69-7·85). Vaccines were well tolerated; reactogenicity was predominantly mild and transient. The percentage of subjects with unsolicited adverse events (AEs) during 28 days after vaccinations was similar among treatments (COVIVAC 3 μg 29·0 %, COVIVAC 6 μg 23·2 %, VAXZEVRIA 31·2 %); no vaccine-related AE was reported. Considering that induction of neutralising antibodies against SARS-CoV-2 has been correlated with the efficacy of COVID-19 vaccines, including VAXZEVRIA, our results suggest that vaccination with COVIVAC may afford clinical benefit matching or exceeding that of the VAXZEVRIA vaccine. ClinicalTrials.govNCT05940194.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Female
- Humans
- Male
- Middle Aged
- Young Adult
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- COVID-19/prevention & control
- COVID-19/immunology
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/adverse effects
- Immunogenicity, Vaccine
- Newcastle disease virus/immunology
- Newcastle disease virus/genetics
- SARS-CoV-2/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Vaccines, Inactivated/immunology
- Vaccines, Inactivated/administration & dosage
- Vaccines, Inactivated/adverse effects
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/adverse effects
- Vaccines, Synthetic/administration & dosage
- Vietnam
Collapse
Affiliation(s)
- Vu Dinh Thiem
- National Institute of Hygiene and Epidemiology, 1 Yersin Street, Hai Ba Trung District, Hanoi, Viet Nam
| | - Dang Duc Anh
- National Institute of Hygiene and Epidemiology, 1 Yersin Street, Hai Ba Trung District, Hanoi, Viet Nam
| | - Vu Hai Ha
- National Institute of Hygiene and Epidemiology, 1 Yersin Street, Hai Ba Trung District, Hanoi, Viet Nam
| | - Nguyen Van Thom
- Center for Disease Control, Thai Binh Province, 10 Hoàng Công Chất street, Quang Trung ward, Thai Binh, Viet Nam
| | - Tran Cong Thang
- PATH Vietnam, Hanoi Towers, 49 Hai Ba Trung Street, Hoan Kiem District, Hanoi, Viet Nam
| | - Jose Mateus
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
| | - Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP) at Mount Sinai New York, New York, NY, USA
| | - Rama Raghunandan
- Center for Vaccine Innovation and Access, PATH, 2201 Westlake Avenue, Suite 200, Seattle, WA 98121, USA.
| | - Nguyen Mai Huong
- PATH Vietnam, Hanoi Towers, 49 Hai Ba Trung Street, Hoan Kiem District, Hanoi, Viet Nam
| | - Laina D Mercer
- Center for Vaccine Innovation and Access, PATH, 2201 Westlake Avenue, Suite 200, Seattle, WA 98121, USA
| | - Jorge Flores
- Center for Vaccine Innovation and Access, PATH, 2201 Westlake Avenue, Suite 200, Seattle, WA 98121, USA
| | - E Alexandar Escarrega
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
| | - Ariel Raskin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP) at Mount Sinai New York, New York, NY, USA
| | - Duong Huu Thai
- Institute of Vaccines and Medical Biologicals, 9 Pasteur, Xuong Huan, Nha Trang City, Khanh Hoa, Viet Nam
| | - Le Van Be
- Institute of Vaccines and Medical Biologicals, 9 Pasteur, Xuong Huan, Nha Trang City, Khanh Hoa, Viet Nam
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA; Department of Medicine, University of California, San Diego, (UCSD), La Jolla, CA, 92037, USA
| | - Bruce L Innis
- Center for Vaccine Innovation and Access, PATH, 2201 Westlake Avenue, Suite 200, Seattle, WA 98121, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP) at Mount Sinai New York, New York, NY, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, NY, New York, USA.
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA; Department of Medicine, University of California, San Diego, (UCSD), La Jolla, CA, 92037, USA.
| |
Collapse
|
49
|
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
|
50
|
Wang X, Kong H, Chu B, Yang Q, Lin C, Liu R, Chen C, Gao Y, Wang G, Wang D, Qin C, Ye X, Yu L, Xu X, Jin J, Sun R, Chen H, Wu X, Zhang Z. Identification of a broad-inhibition influenza neuraminidase antibody from pre-existing memory B cells. Cell Host Microbe 2025; 33:151-166.e8. [PMID: 39740671 DOI: 10.1016/j.chom.2024.12.004] [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: 08/02/2024] [Revised: 09/27/2024] [Accepted: 12/03/2024] [Indexed: 01/02/2025]
Abstract
Identifying broadly reactive B precursor cells and conserved epitopes is crucial for developing a universal flu vaccine. In this study, using influenza neuraminidase (NA) mutant probes, we find that human pre-existing NA-specific memory B cells (MBCs) account for ∼0.25% of total MBCs, which are heterogeneous and dominated by class-unswitched MBCs. In addition, we identify three NA broad-inhibition monoclonal antibodies (mAbs) (BImAbs) that block the activity of NA derived from different influenza strains, including the recent cow H5N1. The cryoelectron microscopy (cryo-EM) structure shows that the BImAb targets the conserved NA enzymatic pocket and a separate epitope in the neighboring NA monomer. Furthermore, the NA BImAbs protect mice from the lethal challenge of the human pandemic H1N1 and H5N1. Our work demonstrates that the NA broad-inhibition precursor MBCs exist in healthy adults and could be targeted by the NA-based universal flu vaccine.
Collapse
Affiliation(s)
- Xin Wang
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Huihui Kong
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Bingxin Chu
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Qian Yang
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Chaohui Lin
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Rui Liu
- School of Life Science, Westlake University, Hangzhou, Zhejiang, China
| | - Changxu Chen
- School of Life Science, Westlake University, Hangzhou, Zhejiang, China
| | - Yang Gao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Guojun Wang
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Dayan Wang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China; WHO Collaborating Center for Reference and Research on Influenza, Beijing, China
| | - Chen Qin
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xiaohua Ye
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Lifei Yu
- Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Xiangfei Xu
- Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Jie Jin
- Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Ren Sun
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China; School of Life Science, Westlake University, Hangzhou, Zhejiang, China
| | - Hualan Chen
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Xudong Wu
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| | - Zeli Zhang
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China; School of Life Science, Westlake University, Hangzhou, Zhejiang, China.
| |
Collapse
|