1
|
Jiang J, Lam KF, Lau EHY, Yin G, Lin Y, Cowling BJ. Protection and waning of vaccine-induced, natural and hybrid immunity to SARS-CoV-2 in Hong Kong. Expert Rev Vaccines 2025; 24:252-260. [PMID: 40137440 DOI: 10.1080/14760584.2025.2485252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 03/24/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND As the COVID-19 pandemic transitions into its fourth year, understanding the dynamics of immunity is critical for implementing effective public health measures. This study examines vaccine-induced, natural, and hybrid immunity to SARS-CoV-2 in Hong Kong, focusing on their protective effectiveness and waning characteristics against infection during the Omicron BA.1/2 dominant period. RESEARCH DESIGN AND METHODS We conducted a territory-wide retrospective cohort study using vaccination and infection records from the Hong Kong Department of Health. The analysis included over 6.5 million adults, applying the Andersen-Gill model to estimate protective effectiveness while addressing selection bias through inverse probability weighting. RESULTS Vaccine-induced immunity peaked one month after the first dose but waned rapidly, while boosters significantly prolonged protection. Infection-induced immunity showed higher initial effectiveness but declined faster than vaccine-induced immunity. Hybrid immunity provided the most durable protection. mRNA vaccines (Comirnaty) demonstrated greater effectiveness and slower waning compared to inactivated vaccines (CoronaVac). CONCLUSIONS Hybrid immunity represents the most effective strategy for sustained protection against SARS-CoV-2. Public health policies should emphasize booster campaigns and hybrid immunity pathways to enhance population-level immunity and guide future COVID-19 management in Hong Kong.
Collapse
Affiliation(s)
- Jialiang Jiang
- Department of Statistics and Actuarial Science, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kwok Fai Lam
- Department of Statistics and Actuarial Science, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore, Singapore
| | - Eric Ho Yin Lau
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
| | - Guosheng Yin
- Department of Statistics and Actuarial Science, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yun Lin
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
| | - Benjamin John Cowling
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
2
|
Purcell RA, Aurelia LC, Allen LF, Bond KA, Williamson DA, Trevillyan JM, Trubiano JA, Wines BD, Hogarth PM, Juno JA, Wheatley AK, Nguyen THO, Subbarao K, Kedzierska K, Kent SJ, Mahanty S, Selva KJ, Chung AW. Genetic markers of enhanced functional antibody responses to COVID-19 vaccination. Vaccine 2025; 61:127379. [PMID: 40527060 DOI: 10.1016/j.vaccine.2025.127379] [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: 12/23/2024] [Accepted: 06/02/2025] [Indexed: 06/19/2025]
Abstract
INTRODUCTION Substantial population-level variation in vaccine-specific antibody responses has been observed following global coronavirus disease 2019 (COVID-19) vaccination efforts. Beyond the influence of clinical and demographic features, immunogenetic variation is suggested to underlie divergent serological responses following COVID-19 vaccination of distinct populations. METHODS Immunoglobulin G1 (IgG1) allotypic markers (G1m) for 121 COVID-19 vaccinated healthy adults were genotyped via Sanger sequencing. Vaccine-specific IgG and Fc gamma receptor (FcγR) engagement were characterised via bead-based multiplex array. RESULTS Following two COVID-19 vaccine doses, G1m1,17+/+ compared to G1m-1,3+/+ vaccinees had increased IgG and FcγR engagement specific for the antigenically conserved SARS-CoV-2 Spike 2 (S2) domain. IgG targeting antigenically novel SARS-CoV-2 receptor binding domain (RBD) trended higher in G1m1,17+/+ vaccinees, facilitating increased RBD-specific FcγR2a-R131 and FcγR2b binding. CONCLUSION Primary COVID-19 vaccination induced increased S2-specific IgG in G1m1,17+/+ vaccinees, facilitating enhanced anti-viral FcγR engagement and suggesting immunogenetics may be a valuble consideration for next-generation vaccine design.
Collapse
Affiliation(s)
- Ruth A Purcell
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| | - L Carissa Aurelia
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Lilith F Allen
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Katherine A Bond
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia; Victorian Infectious Diseases Reference Laboratory (VIDRL), The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Deborah A Williamson
- Department of Infectious Diseases, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia; School of Medicine, University of St Andrews, Fife, KY16 9TF, Scotland
| | - Janine M Trevillyan
- Department of Infectious Diseases, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia; Centre for Antibiotic Allergy and Research, Austin Health, Heidelberg, VIC 3084, Australia
| | - Jason A Trubiano
- Centre for Antibiotic Allergy and Research, Austin Health, Heidelberg, VIC 3084, Australia; Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia; Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Bruce D Wines
- Immune Therapies Group, Burnet Institute, Melbourne, VIC 3004, Australia; Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - P Mark Hogarth
- Immune Therapies Group, Burnet Institute, Melbourne, VIC 3004, Australia; Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Clinical Pathology, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Jennifer A Juno
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Thi H O Nguyen
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Kanta Subbarao
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia; WHO Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia; Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Health, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Siddhartha Mahanty
- Department of Infectious Diseases, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Kevin John Selva
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Amy W Chung
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia.
| |
Collapse
|
3
|
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
|
4
|
Adhikari B, Oltz EM, Gumina RJ, Kick MK, Saif LJ, Vlasova AN. Circulating Antibodies Against Common Cold Coronaviruses Do Not Interfere with Immune Responses to Primary or Booster SARS-CoV-2 mRNA Vaccines. Vaccines (Basel) 2025; 13:547. [PMID: 40432156 PMCID: PMC12115401 DOI: 10.3390/vaccines13050547] [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: 03/31/2025] [Revised: 05/01/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Pre-existing cross-reactive antibodies (Abs) against common cold coronaviruses (CCCoVs) have been hypothesized to influence the immune responses to SARS-CoV-2 vaccine-induced Ab responses. METHODS Serum samples from healthy healthcare workers (HCWs, n = 64) receiving mRNA vaccines were collected at seven time points: pre-COVID-19-vaccination (Pre), post-first dose (Vax1), post-second dose (Vax2), and 6-, 9-, 12-, and 15-months post-Vax2. Booster vaccine doses (n = 23) were received 1-80 days prior to the 9 m sample collection time point. We used peptide-based enzyme-linked immunosorbent assays (ELISAs) to measure SARS-CoV-2/CCCoV-specific IgG/IgA/IgM and SARS-CoV-2 IgG4 (associated with immune tolerance) Ab levels in the HCW serum samples. Additionally, we measured Epstein-Barr/influenza A (unrelated pathogens) virus-specific IgG Ab levels. RESULTS We observed that vaccination significantly increased SARS-CoV-2 IgG Ab levels at the Vax1 (p ≤ 0.0001) and Vax2 (p ≤ 0.0001) time points compared to Pre-Vax. These Ab levels declined at 6 months post-vaccination but increased again following the booster vaccine dose around the 9-month post-Vax2 time point in a cohort (n = 23) of the HCWs. However, this increase was modest compared to those induced by the primary vaccine series. Interestingly, a moderate but continuous increase in SARS-CoV-2 S IgG4 Ab levels was observed throughout this study, becoming statistically significant by the 15-month time point (p = 0.03). Further, a significant increase in CCCoV IgG (but not IgA/IgM) Ab levels was observed at the Vax1 time point, suggestive of cross-reactive or non-specific immune responses. Finally, we observed no negative correlation between the levels of pre-existing CCCoV-specific Abs and the vaccine-induced Ab response (Vax1/Vax2). CONCLUSIONS Pre-existing CCCoV Abs do not interfere with the development of vaccine-induced immunity. However, vaccine-associated Abs wane over time, which may be associated with the increasing IgG4 Ab response.
Collapse
Affiliation(s)
- Bindu Adhikari
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA; (B.A.); (L.J.S.)
- Center for Food Animal Health, Department of Animal Sciences, OARDC, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA;
| | - Eugene M. Oltz
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA;
| | - Richard J. Gumina
- Department of Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH 43210, USA;
| | - Maryssa K. Kick
- Center for Food Animal Health, Department of Animal Sciences, OARDC, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA;
| | - Linda J. Saif
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA; (B.A.); (L.J.S.)
- Center for Food Animal Health, Department of Animal Sciences, OARDC, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA;
| | - Anastasia N. Vlasova
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA; (B.A.); (L.J.S.)
- Center for Food Animal Health, Department of Animal Sciences, OARDC, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA;
| |
Collapse
|
5
|
Jiang J, Lam KF, Lau EHY, Yin G, Lin Y, Cowling BJ. Estimation of trajectory of COVID-19 vaccines effectiveness against infection. Vaccine 2025; 55:127067. [PMID: 40158307 DOI: 10.1016/j.vaccine.2025.127067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/21/2025] [Accepted: 03/21/2025] [Indexed: 04/02/2025]
Abstract
This large-scale cohort study conducted in Hong Kong examined the time-varying protective effects of various COVID-19 vaccines and dosing regimens against the Omicron BA.1/BA.2 variants. An innovative pharmacokinetic/pharmacodynamic model was employed to estimate the trajectory of vaccine effectiveness over time. Results indicated that the maximum protection for a single dose reached 0.120 for CoronaVac and 0.171 for Comirnaty. The peak protective effectiveness for the second and third doses were observed at 0.348 and 0.522, respectively. In a 4-dose regimen, CoronaVac demonstrated a maximum protective effectiveness of 0.548, stabilizing at 0.487, while Comirnaty achieved a maximum effectiveness of 0.784, stabilizing at 0.714 six months after the administration of the last dose. The vaccine effectiveness exhibited a rising and then declining pattern, peaking approximately 1-2 months post-vaccination. Understanding waning immunity is crucial for optimizing vaccination strategies and policies as viral evolution continues. This real-world study captured changing dynamics that may differ from clinical trials with limited follow-up, providing essential evidence to guide the optimization of vaccination efforts. Ongoing monitoring of vaccine effectiveness remains critical as the viral landscape evolves. OBJECTIVES This study aims to investigate the time-varying protective effects of various COVID-19 vaccines and dosing regimens against infections caused by the Omicron BA.1/BA.2 in Hong Kong. METHODS This territory-wide cohort study from Hong Kong combined vaccination records, confirmed COVID-19 cases, and census data from January 2022 to May 2022 to comprehensively analyze the time-varying protective effects of different COVID-19 vaccines and dosing regimens against Omicron BA.1 and BA.2 infections. A 4-parameter pharmacokinetic/pharmacodynamic model was used to estimate the trajectory of vaccine effectiveness over time. RESULTS Among 6.2 million adults, the maximum protective effectiveness for a single vaccine dose reached 0.120 for CoronaVac and 0.171 for Comirnaty. For the second and third doses, peak effectiveness were observed at 0.348 for CoronaVac and 0.522 for Comirnaty. Notably, a 4-dose regimen resulted in maximum protections of 0.548 for CoronaVac and 0.785 for Comirnaty, which stabilized at 0.487 and 0.714, respectively, six months following the last doses. The vaccine effectiveness exhibited a rising then declining pattern, peaking around 1-2 months post-vaccination, underscoring the importance of ongoing vaccination strategies. CONCLUSIONS Understanding the waning of vaccine protection over time is critical for informing optimal vaccination strategies, booster schedules, and public health policies. This real-world study can capture changing dynamics that may differ from clinical trials which have more limited follow-up periods, and can provide crucial evidence to guide optimization of vaccination strategies. Ongoing monitoring of vaccine effectiveness remains crucial as the viral evolution continues.
Collapse
Affiliation(s)
- Jialiang Jiang
- Department of Statistics and Actuarial Science, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region of China
| | - Kwok Fai Lam
- Department of Statistics and Actuarial Science, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region of China; Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore, Singapore.
| | - Eric Ho Yin Lau
- Laboratory of Data Discovery for Health (D(2)4H), Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region of China
| | - Guosheng Yin
- Department of Statistics and Actuarial Science, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region of China
| | - Yun Lin
- Laboratory of Data Discovery for Health (D(2)4H), Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region of China
| | - Benjamin John Cowling
- Laboratory of Data Discovery for Health (D(2)4H), Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region of China; WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region of China
| |
Collapse
|
6
|
Aguareles J, Forné C, García-Casas A, Santamaría-Corral G, Carnevali-Ruiz D, Sotres-Fernández G, Solera JT, Guisado-Vasco P. Influence of anti-Spike protein antibody levels on tocilizumab efficacy in hospitalized patients with severe COVID-19 pneumonia: a post-hoc analysis of the COVACTA trial. BMC Infect Dis 2025; 25:676. [PMID: 40340657 PMCID: PMC12063319 DOI: 10.1186/s12879-025-11001-6] [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: 01/17/2025] [Accepted: 04/17/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Our aim in this work is to find biomarkers to optimize therapy with IL-6 inhibitors, as not all clinical trials have shown clear benefits on mortality or mechanical ventilation progression. Given the link between delayed seroconversion and higher complication risks, we aim to test if evaluating SARS-CoV-2 spike protein antibody status before treatment could enhance IL-6 inhibitor therapy effectiveness in COVID-19 patients. METHODS We conducted a post hoc analysis of the COVACTA study, a phase 3, randomized, double-blind, placebo-controlled trial of the efficacy and safety of tocilizumab in hospitalized patients with severe COVID-19. Cox and logistic regression analysis were used to assess the tocilizumab's efficacy in severe COVID-19 patients on survival and ICU stay at day 28, based on SARS-CoV-2 S-spike and neutralizing antibody levels. RESULTS Tocilizumab reduced 28-day mortality over placebo in patients with low S-spike antibody titers (20% vs. 29%). No benefit was observed for higher antibody levels. Patients with low S-spike antibody levels treated with tocilizumab exhibited a lower probability of ICU stay at day 28 compared to those treated with placebo (63% vs. 82%). No significant differences were noted in mortality and ICU stay based on whole neutralizing antibody titers. CONCLUSIONS Our findings suggest that using IL-6 inhibitors in severe COVID-19 patients with low S-spike antibody titers may improve clinical outcomes. CLINICAL TRIAL Not applicable.
Collapse
Affiliation(s)
- José Aguareles
- Internal Medicine Department, Hospital Universitario Quirónsalud Madrid, Madrid, Spain
- Department of Medicine, Faculty of Medicine, Health and Sports, European University of Madrid, Madrid, Spain
- Research and Clinical Trials Unit, Hospital Universitario Quirónsalud Madrid, Madrid, Spain
| | - Carles Forné
- Heorfy Consulting, Lleida, Spain
- Department of Basic Medical Sciences, University of Lleida, Lleida, Spain
| | - Ana García-Casas
- Internal Medicine Department, Hospital Universitario Quirónsalud Madrid, Madrid, Spain
- Research and Clinical Trials Unit, Hospital Universitario Quirónsalud Madrid, Madrid, Spain
| | - Guillermo Santamaría-Corral
- Internal Medicine Department, Hospital Universitario Quirónsalud Madrid, Madrid, Spain
- Research and Clinical Trials Unit, Hospital Universitario Quirónsalud Madrid, Madrid, Spain
| | - Daniel Carnevali-Ruiz
- Internal Medicine Department, Hospital Universitario Quirónsalud Madrid, Madrid, Spain
- Department of Medicine, Faculty of Medicine, Health and Sports, European University of Madrid, Madrid, Spain
| | - Gabriel Sotres-Fernández
- Internal Medicine Department, Hospital Universitario Quirónsalud Madrid, Madrid, Spain
- Department of Medicine, Faculty of Medicine, Health and Sports, European University of Madrid, Madrid, Spain
| | - Javier T Solera
- Internal Medicine Department, Hospital Universitario Quirónsalud Madrid, Madrid, Spain
- Department of Medicine, Faculty of Medicine, Health and Sports, European University of Madrid, Madrid, Spain
| | - Pablo Guisado-Vasco
- Internal Medicine Department, Hospital Universitario Quirónsalud Madrid, Madrid, Spain.
- Department of Medicine, Faculty of Medicine, Health and Sports, European University of Madrid, Madrid, Spain.
| |
Collapse
|
7
|
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
|
8
|
Kardava L, Lim J, Buckner CM, Lopes de Assis F, Zhang X, Wang W, Melnyk ML, El Merhebi O, Trihemasava K, Teng IT, Carroll R, Jethmalani Y, Castro M, Lin BC, Praiss LH, Seamon CA, Kwong PD, Koup RA, Serebryannyy L, Nickle DC, Chun TW, Moir S. Phenotypic heterogeneity defines B cell responses to repeated SARS-CoV-2 exposures through vaccination and infection. Cell Rep 2025; 44:115557. [PMID: 40222009 PMCID: PMC12080740 DOI: 10.1016/j.celrep.2025.115557] [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: 09/23/2024] [Revised: 01/29/2025] [Accepted: 03/21/2025] [Indexed: 04/15/2025] Open
Abstract
Long-lived humoral memory is key to durable immunity against pathogens yet remains challenging to define due to heterogeneity among antigen-reactive B cells. We addressed this gap through longitudinal sampling over the course of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mRNA vaccinations with or without breakthrough infection. High-dimensional phenotypic profiling performed on ∼72 million B cells showed that receptor-binding domain (RBD) reactivity was associated with five distinct immunoglobulin G (IgG) B cell populations. Two expressed the activation marker CD71, both correlated with neutralizing antibodies, yet the one lacking the memory marker CD27 was induced by vaccination and blunted by infection. Two were resting memory populations; one lacking CD73 arose early and contributed to cross-reactivity; the other, expressing CD73, arose later and correlated with neutralizing antibodies. The fifth, a rare germinal center-like population, contributed to recall responses and was highly cross reactive. Overall, robust and distinct responses to booster vaccination overcame the superiority of hybrid immunity provided by breakthrough infection.
Collapse
Affiliation(s)
- Lela Kardava
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - James Lim
- Monoceros Biosystems, San Diego, CA 29130, USA
| | - Clarisa M Buckner
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Felipe Lopes de Assis
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Xiaozhen Zhang
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Wei Wang
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Mattie L Melnyk
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Omar El Merhebi
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Krittin Trihemasava
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - I-Ting Teng
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Robin Carroll
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | | | - Mike Castro
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Bob C Lin
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Lauren H Praiss
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Catherine A Seamon
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD 20892, USA
| | - Peter D Kwong
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA; Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Richard A Koup
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | | | - David C Nickle
- Monoceros Biosystems, San Diego, CA 29130, USA; Department of Global Health, University of Washington, Seattle, WA 98105, USA
| | - Tae-Wook Chun
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Susan Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
9
|
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
|
10
|
Berends MS, Homburg M, Kupers T, Meijer EN, Bos I, Verheij R, Kuiper J, Berger MY, Peters LL. Impact of pre-existing comorbidities and multimorbidities, demography and viral variants on post-acute sequelae of COVID-19 ('Long COVID') in Dutch primary care: A retrospective cohort study. Int J Infect Dis 2025; 156:107912. [PMID: 40258532 DOI: 10.1016/j.ijid.2025.107912] [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: 03/02/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/23/2025] Open
Abstract
INTRODUCTION Post-acute sequelae of COVID-19 (PASC), or Long COVID, involves persistent symptoms following acute infection, posing a global health challenge. While a growing number of studies have investigated potential predictors and risk factors, uncertainties remain regarding their consistency and clinical applicability. This study investigates PASC prevalence, comorbidities, demographics and viral variants using Dutch primary care electronic healthcare records (EHR). METHODS A retrospective cohort study used EHR data from 59 general practices in the Northern Netherlands, including 19,638 SARS-CoV-2 PCR-positive patients from January 1, 2020, to December 31, 2021. PASC was identified via World Health Organization and CDC guidelines, a Dutch Word2Vec model, and clinical assessments. Relative risk (RR) calculations analysed comorbidities, demographics and viral variants. RESULTS PASC prevalence was 5.8% (95% CI: 5.4-6.1%). Comorbidities significantly increasing PASC risk included lung disease (RR: 1.95), cardiovascular disease (RR: 1.73), diabetes (RR: 1.82), kidney disease (RR: 1.98) and mental illness (RR: 1.29). Females and individuals aged ≥45 had increased risk. Multivariate regression revealed higher odds of prolonged PASC for ages 45-59 (adjusted odds ratios [AOR]: 3.02), 60-74 (AOR: 3.25) and 75+ (AOR: 2.44). Combined mental illness and lung disease further increased risk (AOR: 2.55). CONCLUSION Chronic conditions, multimorbidity and demographics significantly influence PASC onset and duration. Targeted interventions may mitigate its long-term impact.
Collapse
Affiliation(s)
- Matthijs S Berends
- Department of Primary and Long-term Care, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Medical Epidemiology, Certe Foundation, Groningen, The Netherlands
| | - Maarten Homburg
- Department of Primary and Long-term Care, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Thijmen Kupers
- Department of Primary and Long-term Care, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Eline N Meijer
- Department of Primary and Long-term Care, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Isabelle Bos
- Nivel, Netherlands Institute for Health Services Research (Nivel), Utrecht, The Netherlands
| | - Robert Verheij
- Nivel, Netherlands Institute for Health Services Research (Nivel), Utrecht, The Netherlands; Department of Social and Behavioral Sciences, Tilburg University, Tranzo, Tilburg, The Netherlands; Dutch National Health Care Institute, Diemen, The Netherlands
| | - Jeroen Kuiper
- Municipal Health Service Groningen, Groningen, The Netherlands
| | - Marjolein Y Berger
- Department of Primary and Long-term Care, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Lilian L Peters
- Department of Primary and Long-term Care, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Amsterdam UMC Location Vrije Universiteit Amsterdam, Midwifery Science, Amsterdam, The Netherlands; Midwifery Academy Amsterdam Groningen, Inholland, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Saha A, Ghosh Roy S, Dwivedi R, Tripathi P, Kumar K, Nambiar SM, Pathak R. Beyond the Pandemic Era: Recent Advances and Efficacy of SARS-CoV-2 Vaccines Against Emerging Variants of Concern. Vaccines (Basel) 2025; 13:424. [PMID: 40333293 PMCID: PMC12031379 DOI: 10.3390/vaccines13040424] [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: 02/17/2025] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 05/09/2025] Open
Abstract
Vaccination has been instrumental in curbing the transmission of SARS-CoV-2 and mitigating the severity of clinical manifestations associated with COVID-19. Numerous COVID-19 vaccines have been developed to this effect, including BioNTech-Pfizer and Moderna's mRNA vaccines, as well as adenovirus vector-based vaccines such as Oxford-AstraZeneca. However, the emergence of new variants and subvariants of SARS-CoV-2, characterized by enhanced transmissibility and immune evasion, poses significant challenges to the efficacy of current vaccination strategies. In this review, we aim to comprehensively outline the landscape of emerging SARS-CoV-2 variants of concern (VOCs) and sub-lineages that have recently surfaced in the post-pandemic years. We assess the effectiveness of existing vaccines, including their booster doses, against these emerging variants and subvariants, such as BA.2-derived sub-lineages, XBB sub-lineages, and BA.2.86 (Pirola). Furthermore, we discuss the latest advancements in vaccine technology, including multivalent and pan-coronavirus approaches, along with the development of several next-generation coronavirus vaccines, such as exosome-based, virus-like particle (VLP), mucosal, and nanomaterial-based vaccines. Finally, we highlight the key challenges and critical areas for future research to address the evolving threat of SARS-CoV-2 subvariants and to develop strategies for combating the emergence of new viral threats, thereby improving preparedness for future pandemics.
Collapse
Affiliation(s)
- Ankita Saha
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA;
| | - Sounak Ghosh Roy
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Naval Medical Research Command, Silver Spring, MD 20910, USA;
| | - Richa Dwivedi
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, TN 37208, USA;
| | - Prajna Tripathi
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10021, USA;
| | - Kamal Kumar
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA;
| | - Shashank Manohar Nambiar
- Division of Hepatology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA;
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| |
Collapse
|
12
|
Dolange V, Slamanig S, Abdeljawad A, Lai TY, Lemus N, Singh G, Carreño JM, Abbad A, Srivastava K, Simon V, Sachithanandham J, Pekosz A, Sullivan D, Krammer F, Sun W, Palese P, González-Domínguez I. A Surrogate Enzyme-Linked Immunosorbent Assay to Select High-Titer Human Convalescent Plasma for Treating Immunocompromised Patients Infected With Severe Acute Respiratory Syndrome Coronavirus 2 Variants of Concern. J Infect Dis 2025; 231:e723-e733. [PMID: 39749487 PMCID: PMC11998579 DOI: 10.1093/infdis/jiae645] [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: 11/07/2024] [Accepted: 12/31/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND The emergence of new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants challenges the treatment of immunocompromised patients against coronavirus disease 2019 (COVID-19). High-titer COVID-19 convalescent plasma (CCP) remains one of the few available therapeutics for these patients. We have revisited the selection of CCP samples and evaluated their efficacy against the Omicron XBB.1.5 variant, the dominant strain in 2023. METHODS A surrogate enzyme-linked immunoassay was reviewed to select CCP samples that ensure a protective level of neutralizing antibodies as the main correlate of protection. Antibody titers were analyzed in 500 serum samples from a population-based serosurvey at Mount Sinai Hospital in early 2023, and the results were validated with CCP samples (collected in 2020-2023) using an immunosuppressed mouse model. RESULTS Using logistic regression modeling, we have redefined high-titer CCP against the new variant in the postpandemic era, where over 97% of the population has natural or vaccine-induced antibodies against earlier SARS-CoV-2 strains. Treatment of immunocompromised mice with two doses (100 μL/dose) of CCP plasma via intraperitoneal injection reduced lung viral titers by 46-fold 3 days post-XBB.1.5 infection. CONCLUSIONS These findings will guide future efforts in selecting high-titer CCP for emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Victoria Dolange
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Stefan Slamanig
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
- Swammerdam Institute for Life Sciences, University of Amsterdam, The Netherlands
| | - Adam Abdeljawad
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Tsoi Ying Lai
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nicholas Lemus
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
- Center for Vaccine Research and Pandemic Preparedness
| | - Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
- Center for Vaccine Research and Pandemic Preparedness
| | - Anass Abbad
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
- Center for Vaccine Research and Pandemic Preparedness
| | - Komal Srivastava
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
- Center for Vaccine Research and Pandemic Preparedness
| | - Viviana Simon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
- Center for Vaccine Research and Pandemic Preparedness
- Department of Pathology, Molecular and Cell-Based Medicine
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jaiprasath Sachithanandham
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Andrew Pekosz
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - David Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
- Center for Vaccine Research and Pandemic Preparedness
- Department of Pathology, Molecular and Cell-Based Medicine
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Austria
| | - Weina Sun
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Peter Palese
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | | |
Collapse
|
13
|
Carreño JM, Lerman B, Singh G, Abbad A, Yellin T, Ehrenhaus J, Fried M, Nardulli JR, Kang HM, Mulder LCF, Gleason C, Srivastava K, PVI study group, Simon V, Krammer F. XBB.1.5 monovalent vaccine induces lasting cross-reactive responses to SARS-CoV-2 variants such as HV.1 and JN.1, as well as SARS-CoV-1, but elicits limited XBB.1.5 specific antibodies. mBio 2025; 16:e0360724. [PMID: 40042313 PMCID: PMC11980561 DOI: 10.1128/mbio.03607-24] [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: 12/03/2024] [Accepted: 01/13/2025] [Indexed: 04/10/2025] Open
Abstract
The evolution of the antibody response to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is impacted by the nature and number of antigenic exposures. First-generation coronavirus disease 2019 (COVID-19) vaccines encoded an ancestral spike protein. Updated bivalent vaccines and breakthrough infections have shaped the intricate diversity of the polyclonal antibody response and specificity of individual antibody clones. We and others previously showed that bivalent vaccines containing the ancestral and Omicron (BA.5) spikes induce high levels of cross-reactive antibodies but undetectable BA.5-specific antibodies in serum. Here, we assessed sera collected before as well as 1 and 3 months following administration of an updated XBB.1.5 monovalent vaccine to individuals with diverse infection and vaccination histories. Vaccination increased neutralization against recent variants of concern, including HV.1, JN.1, and the vaccine-homologous XBB.1.5. Antibody binding and avidity against ancestral and XBB.1.5 antigens significantly increased after vaccination. However, antibody depletion experiments showed that most of the response was cross-reactive to the ancestral spike, and only low levels of XBB.1.5-specific antibodies to the spike or the receptor-binding domain were detected. Importantly, increased antibody levels were still detectable in circulation 3 months post-vaccination and cross-reacted with severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1) as measured by pseudovirus neutralization and binding assays. Overall, our data suggest that the XBB.1.5 monovalent vaccine predominantly elicits a cross-reactive response imprinted by viral spike antigens encountered early during the pandemic.IMPORTANCEUpdated COVID-19 vaccine formulations and SARS-CoV-2 exposure history affect the antibody response to SARS-CoV-2. High titers of antibodies are induced in serum by XBB.1.5 monovalent vaccination. Antibody depletion experiments reveal that the majority of the antibody response is cross-reactive to the ancestral spike, despite vaccination increasing neutralization against recently circulating Omicron variants. Vaccine-induced SARS-CoV-2 antibodies cross-react with SARS-CoV-1 and remain in the bloodstream for at least 3 months after immunization.
Collapse
Affiliation(s)
- Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
| | - Brian Lerman
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
| | - Anass Abbad
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
| | - Temima Yellin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
| | - Jordan Ehrenhaus
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
| | - Miriam Fried
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
| | - Jessica R. Nardulli
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
| | - Hyun Min Kang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
| | - Lubbertus C. F. Mulder
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
- The Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Charles Gleason
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
| | - Komal Srivastava
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
| | - PVI study group
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
- The Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| | - Viviana Simon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
- The Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
14
|
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
|
15
|
Carmola LR, Roebling AD, Khosravi D, Langsjoen RM, Bombin A, Bixler B, Reid A, Chen C, Wang E, Lu Y, Zheng Z, Zhang R, Nguyen PV, Arthur RA, Fitts E, Gulick DA, Higginbotham D, Taz A, Ahmed A, Crumpler JH, Kraft C, Lam WA, Babiker A, Waggoner JJ, Openo KP, Johnson LM, Westbrook A, Piantadosi A. Viral and host factors associated with SARS-CoV-2 disease severity in Georgia, USA. PLoS One 2025; 20:e0317972. [PMID: 40168303 PMCID: PMC11960886 DOI: 10.1371/journal.pone.0317972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 01/07/2025] [Indexed: 04/03/2025] Open
Abstract
While SARS-CoV-2 vaccines have shown strong efficacy, the continued emergence of new viral variants raises concerns about the ongoing and future public health impact of COVID-19, especially in locations with suboptimal vaccination uptake. We investigated viral and host factors, including vaccination status, that were associated with SARS-CoV-2 disease severity in a setting with low vaccination rates. We analyzed clinical and demographic data from 1,957 individuals in the state of Georgia, USA, coupled with viral genome sequencing from 1,185 samples. We found no specific mutations associated with disease severity. Compared to those who were unvaccinated, vaccinated individuals experienced less severe SARS-CoV-2 disease, and the effect was similar for both variants. Vaccination within the prior 3-9 months was associated with decreased odds of moderate disease, severe disease, and death. Older age and underlying health conditions, especially immunosuppression and renal disease, were associated with increased disease severity. Overall, this study provides insights into the impact of vaccination status, variants/mutations, and clinical factors on disease severity in SARS-CoV-2 infection when vaccination rates are low. Understanding these associations will help refine and reinforce messaging around the crucial importance of vaccination in mitigating the severity of SARS-CoV-2 disease.
Collapse
Affiliation(s)
- Ludy R. Carmola
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Allison Dorothy Roebling
- Georgia Department of Health, Georgia Emerging Infections Program, Atlanta, Georgia, United States of America
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States of America
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Dara Khosravi
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Rose M. Langsjoen
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Andrei Bombin
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Bri Bixler
- Graduate Program in Genetics and Molecular Biology, Emory University, Atlanta, Georgia, United States of America
| | - Alex Reid
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Cara Chen
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Ethan Wang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Yang Lu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Ziduo Zheng
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia, United States of America
| | - Rebecca Zhang
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia, United States of America
| | - Phuong-Vi Nguyen
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Robert A. Arthur
- Emory Integrated Computational Core, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Eric Fitts
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Dalia Arafat Gulick
- Georgia Clinical & Translational Science Alliance, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Dustin Higginbotham
- Georgia Clinical & Translational Science Alliance, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Azmain Taz
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Alaa Ahmed
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Emory Integrated Genomics Core, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - John Hunter Crumpler
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Colleen Kraft
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Wilbur A. Lam
- The Atlanta Center for Microsystems-Engineered Point-of-Care Technologies, Atlanta, Georgia, United States of America
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America,
- Aflac Cancer and Blood Disorders Center at Children’s Healthcare of Atlanta, Atlanta, Georgia, United States of America
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Ahmed Babiker
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Jesse J. Waggoner
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Kyle P. Openo
- Georgia Department of Health, Georgia Emerging Infections Program, Atlanta, Georgia, United States of America
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States of America
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Laura M. Johnson
- Department of Pediatrics, Pediatric Biostatistics Core, School of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Adrianna Westbrook
- Department of Pediatrics, Pediatric Biostatistics Core, School of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Anne Piantadosi
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, United States of America
| |
Collapse
|
16
|
Erice A, Nuño N, Prieto L, Caballero C. Immune Imprinting, Non-Durable Hybrid Immunity, and Hybrid Immune Damping Following SARS-CoV-2 Primary Vaccination with BNT162b2 and Boosting with mRNA-1273. Vaccines (Basel) 2025; 13:310. [PMID: 40266217 PMCID: PMC11945725 DOI: 10.3390/vaccines13030310] [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: 01/19/2025] [Revised: 03/09/2025] [Accepted: 03/10/2025] [Indexed: 04/24/2025] Open
Abstract
Background/Objectives: Long-term studies on the immune response following multiple doses of SARS-CoV-2 mRNA vaccines remain limited. Methods: Secondary analyses of data from a cohort of non-immunocompromised subjects who received two doses of BNT162b2 (primary vaccination) and a booster with mRNA-1273 nine months later. Antibodies targeting the receptor-binding domain of the S1 subunit of the SARS-CoV-2 spike (anti-RBD) were measured at eight time points during follow-up; the SARS-CoV-2-specific T cell response was measured 16 and 25 months after primary vaccination using an interferon-γ release assay. Results: During the 9-month follow up period after primary vaccination and before the mRNA-1273 booster, anti-RBD were significantly higher at all time points in subjects with documented SARS-CoV-2 infection before the first study time point (previously infected subjects; n = 50) compared to naïve subjects (n = 208; p < 0.05). During a 16-month follow up period following the mRNA-1273 booster, anti-RBD were lower at all time points in previously infected subjects (n = 21) compared to naïve subjects (n = 109), although the differences were non-significant. Breakthrough SARS-CoV-2 infections increased over time in both groups, particularly after the mRNA-1273 booster. Most participants had a persistent SARS-CoV-2 specific T cell response regardless of prior infection. Conclusions: These findings suggest a modulating effect of previous SARS-CoV-2 infection on the humoral immune response to mRNA vaccination, a non-durable hybrid immunity following mRNA vaccination in previously infected subjects, and attenuation of the humoral immune response (immune damping) after repeated exposure to SARS-CoV-2 antigens through mRNA vaccination and/or infection.
Collapse
Affiliation(s)
- Alejo Erice
- Department of Internal Medicine, Hospital Asepeyo, 28823 Coslada, Spain
- Facultad de Medicina, Universidad Francisco de Vitoria, 28223 Pozuelo de Alarcón, Spain
| | - Néstor Nuño
- Independent Researcher, 28400 Madrid, Spain;
| | - Lola Prieto
- Unidad de Apoyo a la Investigación, Facultad de Medicina, Universidad Francisco de Vitoria, 28223 Pozuelo de Alarcón, Spain;
| | - Cristina Caballero
- Facultad de Medicina, Universidad Francisco de Vitoria, 28223 Pozuelo de Alarcón, Spain
- Clinical Diagnostic Laboratory, Hospital Asepeyo, 28823 Coslada, Spain;
| |
Collapse
|
17
|
Ao D, Peng D, He C, Ye C, Hong W, Huang X, Lu Y, Shi J, Zhang Y, Liu J, Wei X, Wei Y. A promising mRNA vaccine derived from the JN.1 spike protein confers protective immunity against multiple emerged Omicron variants. MOLECULAR BIOMEDICINE 2025; 6:13. [PMID: 40035925 PMCID: PMC11880457 DOI: 10.1186/s43556-025-00258-7] [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: 12/06/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/06/2025] Open
Abstract
Despite the declared end of the COVID-19 pandemic, SARS-CoV-2 continues to evolve, with emerging JN.1-derived subvariants (e.g., KP.2, KP.3) compromising the efficacy of current XBB.1.5-based vaccines. To address this, we developed an mRNA vaccine encoding the full-length spike protein of JN.1, incorporating GSAS and 2P mutations and encapsulated in lipid nanoparticles (LNPs). The JN.1-mRNA vaccine elicited robust humoral and cellular immune responses in mice, including high JN.1-specific IgG titers, cross-neutralizing antibodies, and increased T follicular helper (Tfh) cells, germinal center (GC) B cells, and T cell cytokines. Importantly, immunity persisted for up to six months and induced RBD-specific long-lived plasma cells. We also compared the immune responses induced by homologous and heterologous vaccination regimens, and our results demonstrated that the heterologous regimen-combining JN.1-mRNA with a recombinant protein vaccine (RBDJN.1-HR)-induced stronger responses. These findings highlight the JN.1-mRNA vaccine constitutes an effective prophylactic approach against JN.1-related variants, as it induces potent neutralizing antibody responses across all tested lineages. This enhanced immunogenicity is expected to significantly reduce hospitalization rates and mitigate post-COVID complications associated with JN.1 and KP.3 infections. This study emphasizes the need for timely vaccine updates and the adaptability of mRNA vaccines in addressing emerging pathogens, providing a framework for combating future infectious diseases. Collectively, these results offer critical insights for vaccine design and public health strategies in response to emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Danyi Ao
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China
| | - Dandan Peng
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China
| | - Cai He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China
| | - Chunjun Ye
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China
| | - Xiya Huang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China
| | - Yishan Lu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China
| | - Jie Shi
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China
| | - Yu Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China
| | - Jian Liu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China.
- WestVac Biopharma Co. Ltd., Chengdu, China.
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
18
|
Kang HM, Kim HJ, Jung J, Ahn JY, Song KH, Baek JY, Choi JY, Lee YJ, Jeong H, Kim SH, Park S, Jang HM, Rhie GE, Kim ES, Choi JY, Kim SH, Kang ES, Peck KR, Jeong HW, Ko JH. Natural Boosting and the Immunogenicity of the XBB.1.5 Monovalent Vaccine in the Coronavirus Disease 2019 Endemic Era: A Longitudinal Observational Study. J Infect Dis 2025; 231:392-402. [PMID: 39612911 DOI: 10.1093/infdis/jiae536] [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: 08/29/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND With the transition from the coronavirus disease 2019 (COVID-19) pandemic into endemicity, changes in group immunity and the effect of updated XBB.1.5 monovalent vaccine (MonoV) need to be investigated. METHODS A multicenter vaccine cohort was followed for 3 years, and the investigation period was classified into the pre-Omicron, Omicron, and endemic eras. Thirteen sampling points were assessed, including pre- and post-MonoV administration. Specimens were classified as vaccinated, molecularly or serologically diagnosed breakthrough infection (BI), natural boosting (NB), or waned. RESULTS A total of 327 healthcare workers contributed 2645 blood samples from March 2021 to December 2023. The log10 anti-spike protein antibody (SAb) levels, elevated by vaccination, declined linearly in the pre-Omicron era, were maintained during the Omicron era due to BIs, and increased in the endemic era (slope = 0.02, P = .02) without additional vaccination. NB cases increased significantly across the epidemiologic eras. The incidence rate ratios were 2.72 (P < .001) for Omicron/pre-Omicron and 3.39 (P < .001) for endemic/Omicron. Plaque reduction neutralization test (PRNT) titers against circulating strains (XBB.1.5 and XBB.1.9.1) in the NB group maintained previous levels, but ratios to wild-type PRNT and fold changes exhibited significantly enhanced activity. The XBB.1.5 MonoV increased PRNT by 5.8-fold against XBB.1.5 and 6.6-fold against JN.1, showing stronger enhancement against subsequent epidemic strains than the bivalent vaccine. CONCLUSIONS Group immunity in the COVID-19 endemic era exhibited maintained SAb levels and adjusted neutralizing activities through BI and NB. The XBB.1.5 MonoV significantly enhanced neutralizing activity against the vaccine strain and robust immunity against the subsequent epidemic JN.1 strain.
Collapse
Affiliation(s)
- Hyun Myung Kang
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hye-Jin Kim
- Center for Vaccine Research, National Institute of Infectious Diseases, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Jiwon Jung
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin Young Ahn
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyoung-Ho Song
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Jin Yang Baek
- Asia Pacific Foundation for Infectious Diseases (APFID), Seoul, Republic of Korea
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ju-Yeon Choi
- Center for Vaccine Research, National Institute of Infectious Diseases, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Young Jae Lee
- Center for Vaccine Research, National Institute of Infectious Diseases, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Hyeonji Jeong
- Center for Vaccine Research, National Institute of Infectious Diseases, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Su-Hwan Kim
- Center for Vaccine Research, National Institute of Infectious Diseases, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Soyoung Park
- Center for Vaccine Research, National Institute of Infectious Diseases, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Hye Min Jang
- Center for Vaccine Research, National Institute of Infectious Diseases, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Gi-Eun Rhie
- Center for Vaccine Research, National Institute of Infectious Diseases, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Eu Suk Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Jun Yong Choi
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sung-Han Kim
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eun-Suk Kang
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyong Ran Peck
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hye Won Jeong
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
| | - Jae-Hoon Ko
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
19
|
Rubio R, Macià D, Barrios D, Vidal M, Jiménez A, Molinos-Albert LM, Díaz N, Canyelles M, Lara-Escandell M, Planchais C, Santamaria P, Carolis C, Izquierdo L, Aguilar R, Moncunill G, Dobaño C. High-resolution kinetics and cellular determinants of SARS-CoV-2 antibody response over two years after COVID-19 vaccination. Microbes Infect 2025; 27:105423. [PMID: 39299570 DOI: 10.1016/j.micinf.2024.105423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/07/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) studies usually rely on cross-sectional data of large cohorts but limited repeated samples, overlooking significant inter-individual antibody kinetic differences. By combining Luminex, activation-induced marker (AIM) and IFN-γ/IL-2 Fluorospot assays, we characterized the IgM, IgA, and IgG antibody kinetics using 610 samples from 31 healthy adults over two years after COVID-19 vaccination, and the T-cell responses six months post-booster. Antibody trajectories varied among isotypes: IgG decayed slowly, IgA exhibited an initial sharp decline, which gradually slowed down and stabilized above the seropositivity threshold. Contrarily, IgM rapidly dropped to undetectable levels after primary vaccination. Importantly, three vaccine doses induced higher and more durable anti-spike IgG and IgA levels compared to two doses, whereas infection led to the highest antibody peak and slowest antibody decay rate compared to vaccination. Comparing with ancestral virus, antibody levels recognizing Omicron subvariants had a faster antibody decay. Finally, polyfunctional T cells were positively associated with subsequent IgA responses. These results revealed distinctive antibody patterns by isotype and highlight the benefits of booster doses in enhancing and sustaining antibody responses.
Collapse
Affiliation(s)
- Rocío Rubio
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
| | - Dídac Macià
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Diana Barrios
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
| | - Marta Vidal
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
| | - Alfons Jiménez
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain; CIBER de Salud Pública y Epidemiología (CIBERESP), Barcelona, Spain
| | - Luis M Molinos-Albert
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
| | - Natalia Díaz
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
| | - Mar Canyelles
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
| | - Maria Lara-Escandell
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
| | - Cyril Planchais
- Laboratory of Humoral Immunology, Institut Pasteur, Université Paris Cité, F-75015 Paris, France
| | - Pere Santamaria
- Pathogenesis and Treatment of Autoimmunity Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Carlo Carolis
- Biomolecular Screening and Protein Technologies Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Luis Izquierdo
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Ruth Aguilar
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
| | - Gemma Moncunill
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain.
| | - Carlota Dobaño
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain.
| |
Collapse
|
20
|
Herrera BB, Chaplin B, MBoup S, Abdullahi A, He M, Fisher SM, Akanmu S, Chang CA, Hamel DJ, Gupta RK, Kanki PJ. Pre-pandemic cross-reactive antibody and cellular responses against SARS-CoV-2 among female sex workers in Dakar, Senegal. Front Public Health 2025; 13:1522733. [PMID: 39916712 PMCID: PMC11798920 DOI: 10.3389/fpubh.2025.1522733] [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: 11/26/2024] [Accepted: 01/06/2025] [Indexed: 02/09/2025] Open
Abstract
Background The COVID-19 pandemic had a severe impact globally, yet African populations exhibited unexpectedly lower rates of severe disease and mortality. We investigated the potential role of pre-existing immunity in shaping the epidemiology of COVID-19 in Africa. Methods Plasma collected from Senegalese female sex workers prior to the COVID-19 pandemic was screened for SARS-CoV-2 and human coronavirus (hCoV) antibodies by virion immunoblots. For antibody-reactive plasma, paired peripheral blood mononuclear cells were stimulated by fusion proteins and IFN-γ cellular responses were assessed via ELISPOT. Results We observed substantial levels of pre-existing cross-reactive immunity to SARS-CoV-2, stemming from prior exposure to seasonal hCoVs. Our antibody analysis revealed a 23.5% (47/200) seroprevalence rate against SARS-CoV-2 nucleocapsid (N). These samples were then probed for antibodies against hCoV spike (S) and/or N antigens; 85.1% (40/47), 70.2% (33/47), and 95.7% (45/47) were antibody reactive against hCoV-229E, hCoV-OC43, or hCoV-HKU1, respectively. Our analysis of cellular responses also demonstrated cross-reactivity to SARS-CoV-2 with 80.0% (36/45) and 82.2% (37/45) showing IFN-γ responses against S and N, respectively. A unique pre-pandemic subject had cross-reactive SARS-CoV-2 S antibodies with detectable neutralization and cross-reactive cellular responses. Conclusion These findings suggest that prior hCoV exposure may induce cross-reactive adaptive immunity, potentially contributing to protection against COVID-19. Our study provides unique data on the dynamics of hCoV and SARS-CoV-2 immunity in Senegal and underscores the importance of understanding the role of pre-existing immunity in shaping COVID-19 outcomes globally.
Collapse
Affiliation(s)
- Bobby Brooke Herrera
- Rutgers Global Health Institute, Rutgers University, New Brunswick, NJ, United States
- Department of Medicine, Division of Allergy, Immunology, and Infectious Diseases, and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, United States
- Mir Biosciences, Inc., Dunellen, NJ, United States
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Beth Chaplin
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Souleymane MBoup
- Institut De Recherche En Santé De Surveillance Épidémiologique Et De Formation (IRESSEF), Dakar, Senegal
| | - Adam Abdullahi
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Michelle He
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Sydney M. Fisher
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Sulaimon Akanmu
- Lagos University Teaching Hospital, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Charlotte A. Chang
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Donald J. Hamel
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Ravindra K. Gupta
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Phyllis J. Kanki
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| |
Collapse
|
21
|
Hou CW, Williams S, Boyle V, Roeder A, Bobbett B, Garcia I, Caruth G, Magee M, Chung Y, Lake DF, LaBaer J, Murugan V. Tracking Immunity: An Increased Number of COVID-19 Boosters Increases the Longevity of Anti-RBD and Anti-RBD-Neutralizing Antibodies. Vaccines (Basel) 2025; 13:61. [PMID: 39852840 PMCID: PMC11769131 DOI: 10.3390/vaccines13010061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES Since the World Health Organization declared COVID-19 a pandemic in March 2020, the virus has caused multiple waves of infection globally. Arizona State University (ASU), the largest four-year university in the United States, offers a uniquely diverse setting for assessing immunity within a large community. This study aimed to test our hypothesis that an increased number of exposures to SARS-CoV-2 RBD through vaccination/boosters/infection will increase SARS-CoV-2 antibody seroprevalence by increasing the longevity of anti-RBD and anti-RBD-neutralizing antibodies. METHODS A serosurvey was conducted at ASU from 30 January to 3 February 2023. Participants completed questionnaires about demographics, respiratory infection history, symptoms, and COVID-19 vaccination status. Blood samples were analyzed for anti-receptor binding domain (RBD) IgG and anti-nucleocapsid (NC) antibodies, offering a comprehensive view of immunity from both natural infection and vaccination. RESULTS The seroprevalence of anti-RBD IgG antibodies was 96.2% (95% CI: 94.8-97.2%), and 64.9% (95% CI: 61.9-67.8%) of participants had anti-NC antibodies. Anti-RBD IgG levels correlated strongly with neutralizing antibody levels, and participants who received more vaccine doses showed higher levels of both anti-RBD IgG and neutralizing antibodies. Increasing the number of exposures through vaccination and/or infection resulted in higher and long-lasting antibodies. CONCLUSIONS The high levels of anti-RBD antibodies observed reflect substantial vaccine uptake within this population. Ongoing vaccination efforts, especially as new variants emerge, are essential to maintaining protective antibody levels. These findings underscore the importance of sustained public health initiatives to support broad-based immunity and protection.
Collapse
Affiliation(s)
- Ching-Wen Hou
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (C.-W.H.); (S.W.); (V.B.); (B.B.); (I.G.); (G.C.); (M.M.); (Y.C.); (J.L.)
| | - Stacy Williams
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (C.-W.H.); (S.W.); (V.B.); (B.B.); (I.G.); (G.C.); (M.M.); (Y.C.); (J.L.)
| | - Veronica Boyle
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (C.-W.H.); (S.W.); (V.B.); (B.B.); (I.G.); (G.C.); (M.M.); (Y.C.); (J.L.)
| | - Alexa Roeder
- School of Life Sciences, Arizona State University, Phoenix, AZ 85004, USA; (A.R.); (D.F.L.)
| | - Bradley Bobbett
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (C.-W.H.); (S.W.); (V.B.); (B.B.); (I.G.); (G.C.); (M.M.); (Y.C.); (J.L.)
| | - Izamar Garcia
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (C.-W.H.); (S.W.); (V.B.); (B.B.); (I.G.); (G.C.); (M.M.); (Y.C.); (J.L.)
| | - Giavanna Caruth
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (C.-W.H.); (S.W.); (V.B.); (B.B.); (I.G.); (G.C.); (M.M.); (Y.C.); (J.L.)
| | - Mitch Magee
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (C.-W.H.); (S.W.); (V.B.); (B.B.); (I.G.); (G.C.); (M.M.); (Y.C.); (J.L.)
| | - Yunro Chung
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (C.-W.H.); (S.W.); (V.B.); (B.B.); (I.G.); (G.C.); (M.M.); (Y.C.); (J.L.)
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
| | - Douglas F. Lake
- School of Life Sciences, Arizona State University, Phoenix, AZ 85004, USA; (A.R.); (D.F.L.)
| | - Joshua LaBaer
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (C.-W.H.); (S.W.); (V.B.); (B.B.); (I.G.); (G.C.); (M.M.); (Y.C.); (J.L.)
- School of Molecular Sciences, Arizona State University, Phoenix, AZ 85004, USA
| | - Vel Murugan
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (C.-W.H.); (S.W.); (V.B.); (B.B.); (I.G.); (G.C.); (M.M.); (Y.C.); (J.L.)
| |
Collapse
|
22
|
Hou CW, Williams S, Trivino-Soto G, Boyle V, Rainford D, Vicino S, Magee M, Chung Y, LaBaer J, Murugan V. The omicron variant of SARS-CoV-2 drove broadly increased seroprevalence in a public university setting. PLOS GLOBAL PUBLIC HEALTH 2025; 5:e0003893. [PMID: 39752417 PMCID: PMC11698426 DOI: 10.1371/journal.pgph.0003893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 10/03/2024] [Indexed: 01/06/2025]
Abstract
Omicron is the comparatively most transmissible and contagious variant of severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2). We conducted a seroprevalence study from March 1-3, 2022, to investigate the seroprevalence of SARS-CoV-2 antibodies among individuals aged 18 years and older after the Omicron outbreak. The seroprevalence of anti-receptor binding domain (RBD) antibodies was found to be 96.3% (95% CI 95.2-97.2%) compared to 88.2% (95% CI 86.1-90%) in our previous serosurvey. For anti-nucleocapsid (NC) antibodies, the seroprevalence was 39.1% (95% CI 36.6-41.7%) compared to 19.7% (95% CI 17.5-22.2%) earlier. Individuals that experienced breakthrough infections exhibited the highest levels of anti-RBD antibodies. Additionally, saliva samples showed promise as a potential diagnostic biofluid for measuring antibody levels, as they exhibited a strong agreement with the data obtained from serum samples. The near doubling of anti-NC reactivity, a proxy for history of infection, reflects the contagiousness of the omicron variant, but may also have been influenced by a more relaxed approach to precautions in the spring of 2022. Serosurveys repeated at regular intervals monitor the trend of infections in the community, delineate the geographical spread of the infection, and may guide containment measures in communities, and prompt response to future outbreaks.
Collapse
Affiliation(s)
- Ching-Wen Hou
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Stacy Williams
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Guillermo Trivino-Soto
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Veronica Boyle
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - David Rainford
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Selina Vicino
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Mitch Magee
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Yunro Chung
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
- College of Health Solutions, Arizona State University, Phoenix, Arizona, United States of America
| | - Joshua LaBaer
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Vel Murugan
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| |
Collapse
|
23
|
De Oliveira AS, Versteeg L, Briggs N, Adhikari R, Villar MJ, Redd JR, Hotez P, Bottazzi ME, Pollet J. Altering the intracellular trafficking of Necator americanus GST-1 antigen yields novel hookworm mRNA vaccine candidates. PLoS Negl Trop Dis 2025; 19:e0012809. [PMID: 39792959 PMCID: PMC11756802 DOI: 10.1371/journal.pntd.0012809] [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: 07/12/2024] [Revised: 01/23/2025] [Accepted: 12/24/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND The antigen Na-GST-1, expressed by the hookworm Necator americanus, plays crucial biochemical roles in parasite survival. This study explores the development of mRNA vaccine candidates based on Na-GST-1, building on the success of recombinant Na-GST-1 (rNa-GST-1) protein, currently assessed as a subunit vaccine candidate, which has shown promise in preclinical and clinical studies. METHODOLOGY/FINDINGS By leveraging the flexible design of RNA vaccines and protein intracellular trafficking signal sequences, we developed three variants of Na-GST-1 as native (cytosolic), secretory, and plasma membrane-anchored (PM) antigens. After one immunization in mice, mRNA vaccines induced an earlier onset of antigen-specific antibodies compared to rNa-GST-1. Following two immunizations, mRNA vaccines induced similar or superior levels of antigen-specific antibodies compared to rNa-GST-1. Secretory Na-GST-1 was comparable to rNa-GST1 in producing neutralizing antibodies against Na-GST-1's thiol transferase activity, while native Na-GST-1 induced a more robust CD8+ T cell response due to its intracellular accumulation. Although PM Na-GST-1 elicited one of highest titers of antigen-specific antibody and a diverse set of memory T-cell populations, it resulted in a lower ratio of neutralizing antibodies after IgG purification compared to the other vaccine candidates. CONCLUSIONS/SIGNIFICANCE These findings emphasize the importance of antigen localization in tailoring immune responses and suggest that extracellular antigens are more effective for inducing humoral responses, whereas cytosolic antigen accumulation enhances MHC-1 peptide presentation. Future studies will determine if these in vitro and immunogenicity findings translate to in vivo efficacy. Altogether, mRNA vaccines offer numerous possibilities in the development of multivalent vaccines with single or multiple antigens.
Collapse
Affiliation(s)
- Athos Silva De Oliveira
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children’s Hospital Center for Vaccine Development, Houston, Texas, United States of America
| | - Leroy Versteeg
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children’s Hospital Center for Vaccine Development, Houston, Texas, United States of America
| | - Neima Briggs
- Departments of Immunobiology and Internal Medicine (Infectious Diseases), Yale University, New Haven, Connecticut, United States of America
| | - Rakesh Adhikari
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children’s Hospital Center for Vaccine Development, Houston, Texas, United States of America
| | - Maria Jose Villar
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children’s Hospital Center for Vaccine Development, Houston, Texas, United States of America
| | - JeAnna R. Redd
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children’s Hospital Center for Vaccine Development, Houston, Texas, United States of America
| | - Peter Hotez
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children’s Hospital Center for Vaccine Development, Houston, Texas, United States of America
- Department of Biology, Baylor University, Waco, Texas, United States of America
- James A. Baker III Institute for Public Policy, Rice University, Houston, Texas, United States of America
| | - Maria Elena Bottazzi
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children’s Hospital Center for Vaccine Development, Houston, Texas, United States of America
- Department of Biology, Baylor University, Waco, Texas, United States of America
- James A. Baker III Institute for Public Policy, Rice University, Houston, Texas, United States of America
| | - Jeroen Pollet
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children’s Hospital Center for Vaccine Development, Houston, Texas, United States of America
| |
Collapse
|
24
|
Poletti P. Kinetics of neutralising antibodies against SARS-CoV-2 variants. THE LANCET. INFECTIOUS DISEASES 2025; 25:8-9. [PMID: 39276781 DOI: 10.1016/s1473-3099(24)00513-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 08/05/2024] [Indexed: 09/17/2024]
Affiliation(s)
- Piero Poletti
- Center for Health Emergencies, Fondazione Bruno Kessler, Trento 38123, Italy.
| |
Collapse
|
25
|
Angius F, Puxeddu S, Zaimi S, Canton S, Nematollahzadeh S, Pibiri A, Delogu I, Alvisi G, Moi ML, Manzin A. SARS-CoV-2 Evolution: Implications for Diagnosis, Treatment, Vaccine Effectiveness and Development. Vaccines (Basel) 2024; 13:17. [PMID: 39852796 PMCID: PMC11769326 DOI: 10.3390/vaccines13010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025] Open
Abstract
The COVID-19 pandemic, driven by the rapid evolution of the SARS-CoV-2 virus, presents ongoing challenges to global public health. SARS-CoV-2 is characterized by rapidly evolving mutations, especially in (but not limited to) the spike protein, complicating predictions about its evolutionary trajectory. These mutations have significantly affected transmissibility, immune evasion, and vaccine efficacy, leading to multiple pandemic waves with over half a billion cases and seven million deaths globally. Despite several strategies, from rapid vaccine development and administration to the design and availability of antivirals, including monoclonal antibodies, already having been employed, the persistent circulation of the virus and the emergence of new variants continue to result in high case numbers and fatalities. In the past four years, immense research efforts have contributed much to our understanding of the viral pathogenesis mechanism, the COVID-19 syndrome, and the host-microbe interactions, leading to the development of effective vaccines, diagnostic tools, and treatments. The focus of this review is to provide a comprehensive analysis of the functional impact of mutations on diagnosis, treatments, and vaccine effectiveness. We further discuss vaccine safety in pregnancy and the implications of hybrid immunity on long-term protection against infection, as well as the latest developments on a pan-coronavirus vaccine and nasal formulations, emphasizing the need for continued surveillance, research, and adaptive public health strategies in response to the ongoing SARS-CoV-2 evolution race.
Collapse
Affiliation(s)
- Fabrizio Angius
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Silvia Puxeddu
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Silvio Zaimi
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Serena Canton
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Sepehr Nematollahzadeh
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (S.N.); (G.A.)
| | - Andrea Pibiri
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Ilenia Delogu
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Gualtiero Alvisi
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (S.N.); (G.A.)
| | - Meng Ling Moi
- School of International Health, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Aldo Manzin
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| |
Collapse
|
26
|
Brandi R, Paganelli A, D’Amelio R, Giuliani P, Lista F, Salemi S, Paganelli R. mRNA Vaccines Against COVID-19 as Trailblazers for Other Human Infectious Diseases. Vaccines (Basel) 2024; 12:1418. [PMID: 39772079 PMCID: PMC11680146 DOI: 10.3390/vaccines12121418] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/07/2024] [Accepted: 12/13/2024] [Indexed: 01/03/2025] Open
Abstract
mRNA vaccines represent a milestone in the history of vaccinology, because they are safe, very effective, quick and cost-effective to produce, easy to adapt should the antigen vary, and able to induce humoral and cellular immunity. METHODS To date, only two COVID-19 mRNA and one RSV vaccines have been approved. However, several mRNA vaccines are currently under development for the prevention of human viral (influenza, human immunodeficiency virus [HIV], Epstein-Barr virus, cytomegalovirus, Zika, respiratory syncytial virus, metapneumovirus/parainfluenza 3, Chikungunya, Nipah, rabies, varicella zoster virus, and herpes simplex virus 1 and 2), bacterial (tuberculosis), and parasitic (malaria) diseases. RESULTS RNA viruses, such as severe acute respiratory syndrome coronavirus (SARS-CoV)-2, HIV, and influenza, are characterized by high variability, thus creating the need to rapidly adapt the vaccines to the circulating viral strain, a task that mRNA vaccines can easily accomplish; however, the speed of variability may be higher than the time needed for a vaccine to be adapted. mRNA vaccines, using lipid nanoparticles as the delivery system, may act as adjuvants, thus powerfully stimulating innate as well as adaptive immunity, both humoral, which is rapidly waning, and cell-mediated, which is highly persistent. Safety profiles were satisfactory, considering that only a slight increase in prognostically favorable anaphylactic reactions in young females and myopericarditis in young males has been observed. CONCLUSIONS The COVID-19 pandemic determined a shift in the use of RNA: after having been used in medicine as micro-RNAs and tumor vaccines, the new era of anti-infectious mRNA vaccines has begun, which is currently in great development, to either improve already available, but unsatisfactory, vaccines or develop protective vaccines against infectious agents for which no preventative tools have been realized yet.
Collapse
Affiliation(s)
- Rossella Brandi
- Istituto di Science Biomediche della Difesa, Stato Maggiore Della Difesa, 00184 Rome, Italy; (R.B.); (F.L.)
| | | | | | - Paolo Giuliani
- Poliambulatorio Montezemolo, Ente Sanitario Militare del Ministero Della Difesa Presso la Corte dei Conti, 00195 Rome, Italy;
| | - Florigio Lista
- Istituto di Science Biomediche della Difesa, Stato Maggiore Della Difesa, 00184 Rome, Italy; (R.B.); (F.L.)
| | - Simonetta Salemi
- Division of Internal Medicine, Azienda Ospedaliero-Universitaria S. Andrea, 00189 Rome, Italy
| | - Roberto Paganelli
- Internal Medicine, Faculty of Medicine and Surgery, Unicamillus, International School of Medicine, 00131 Rome, Italy
| |
Collapse
|
27
|
Kusunoki H. Current Status and Significance of Additional Vaccination with COVID-19 Vaccine in Japan-Considerations from Antibody Levels from Hybrid Immunity and Public Perceptions. Vaccines (Basel) 2024; 12:1413. [PMID: 39772074 PMCID: PMC11680318 DOI: 10.3390/vaccines12121413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
This report examines the evolving role of coronavirus disease 2019 (COVID-19) vaccination in Japan, especially in light of the reduced public concern following the reclassification of COVID-19 as a Category 5 infectious disease in May 2023. With over half the population estimated to have hybrid immunity from prior infections and vaccinations, this report evaluated the necessity and frequency of additional booster doses. Despite strong recommendations from Japanese medical societies to continue vaccination, public skepticism remains owing to financial burdens, adverse reactions, and the perceived limited benefits of frequent boosters. Studies on antibody responses have revealed that individuals with hybrid immunity maintain robust protection with significantly elevated antibody titers that persist over extended periods. Case studies have indicated durable immunity among individuals who have both been vaccinated and experienced breakthrough infections, raising questions about the need for uniform booster policies. This report also discusses the newly approved replicon-type (self-amplifying) vaccines currently available only in Japan, which have generated public and professional debates regarding their efficacy and safety. A more personalized approach to vaccination that takes into account the antibody titers, prior infection history, and individual choices is recommended. Finally, this report underscores the importance of aligning vaccination policies with scientific evidence and public sentiment to optimize COVID-19 countermeasures in Japan.
Collapse
Affiliation(s)
- Hiroshi Kusunoki
- Department of Internal Medicine, Osaka Dental University, 8-1 Kuzuhahanazonocho, Hirakata 573-1121, Osaka, Japan
| |
Collapse
|
28
|
Odle A, Kar M, Verma AK, Sariol A, Meyerholz DK, Suthar MS, Wong LYR, Perlman S. Tissue-resident memory T cells contribute to protection against heterologous SARS-CoV-2 challenge. JCI Insight 2024; 9:e184074. [PMID: 39405115 PMCID: PMC11623939 DOI: 10.1172/jci.insight.184074] [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: 06/18/2024] [Accepted: 10/08/2024] [Indexed: 12/07/2024] Open
Abstract
New vaccine formulations are based on circulating strains of virus, which have tended to evolve to more readily transmit human to human and to evade the neutralizing antibody response. An assumption of this approach is that ancestral strains of virus will not recur. Recurrence of these strains could be a problem for individuals not previously exposed to ancestral spike protein. Here, we addressed this by infecting mice with recent SARS-CoV-2 variants and then challenging them with a highly pathogenic mouse-adapted virus closely related to the ancestral Wuhan-1 strain (SARS2-N501YMA30). We found that challenged mice were protected from severe disease, despite having low or no neutralizing antibodies against SARS2-N501YMA30. T cell depletion from previously infected mice did not diminish infection against clinical disease, although it resulted in delayed virus clearance in the nasal turbinate and, in some cases, in the lungs. Levels of tissue-resident memory T cells were significantly elevated in the nasal turbinate of previously infected mice compared with that of naive mice. However, this phenotype was not seen in lung tissues. Together, these results indicate that the immune response to newly circulating variants afforded protection against reinfection with the ancestral virus that was in part T cell based.
Collapse
Affiliation(s)
- Abby Odle
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Meenakshi Kar
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Vaccine Center, Atlanta, Georgia, USA
| | - Abhishek K. Verma
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Alan Sariol
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Mehul S. Suthar
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Vaccine Center, Atlanta, Georgia, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Lok-Yin Roy Wong
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
- Department of Microbiology, Biochemistry and Molecular Genetics and
- Center for Virus-Host-Innate Immunity, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
29
|
Harrache A, Saker K, Mokdad B, Generenaz L, Saade C, Pons S, Fassier JB, Bal A, Trabaud MA, Rabilloud M, Abichou-Klich A, Trouillet-Assant S. Anti-RBD IgG dynamics following infection or vaccination. Vaccine 2024; 42:126464. [PMID: 39432992 DOI: 10.1016/j.vaccine.2024.126464] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 09/10/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024]
Abstract
Identifying parameters influencing SARS-CoV-2 antibody dynamics post infection or vaccination is crucial for refining vaccination strategies. In a longitudinal analysis of 1340 samples from 375 healthcare workers, we characterized peak serological response and IgG half-life. Peak antibody titers post 2 vaccine doses were ∼ 20 times higher than natural infection; conversely, infected individuals had extended antibody half-life. Clinical and demographical factors such as BMI, age and smoking shaped peak response without affecting anti-RBD IgG half-life. A third mRNA vaccine dose increased peak antibody titers and prolonged half-life compared to the second dose. These findings underscore the diverse kinetics of SARS-CoV-2 antibody responses, which is influenced by immunization type/number and clinical factors.
Collapse
Affiliation(s)
- Amira Harrache
- Biostatistics Department, Hospices Civils de Lyon, Lyon France. CNRS, UMR 5558, University of Lyon, Biometrics and Evolutionary Biology Laboratory, Biostatistics-Health Team, 69100 Villeurbanne, France
| | - Kahina Saker
- Joint Research Unit Hospices Civils of Lyon-bioMérieux, Lyon Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France
| | - Bouchra Mokdad
- Joint Research Unit Hospices Civils of Lyon-bioMérieux, Lyon Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France
| | - Laurence Generenaz
- Joint Research Unit Hospices Civils of Lyon-bioMérieux, Lyon Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France
| | - Carla Saade
- Joint Research Unit Hospices Civils of Lyon-bioMérieux, Lyon Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France
| | - Sylvie Pons
- Joint Research Unit Hospices Civils of Lyon-bioMérieux, Lyon Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France
| | - Jean-Baptiste Fassier
- Occupational Health and Medicine Department, Hospices Civils de Lyon, 69002 Lyon, France
| | - Antonin Bal
- Virology Laboratory, Institute of Infectious Agents, Laboratory associated with the National Reference Centre for Respiratory Infection Viruses, Hospices Civils de Lyon, IAI, North Biology Centre, North Hospital Group, 69004 Lyon, France
| | - Mary-Anne Trabaud
- Virology Laboratory, Institute of Infectious Agents, Laboratory associated with the National Reference Centre for Respiratory Infection Viruses, Hospices Civils de Lyon, IAI, North Biology Centre, North Hospital Group, 69004 Lyon, France
| | - Muriel Rabilloud
- Biostatistics Department, Hospices Civils de Lyon, Lyon France. CNRS, UMR 5558, University of Lyon, Biometrics and Evolutionary Biology Laboratory, Biostatistics-Health Team, 69100 Villeurbanne, France
| | - Amna Abichou-Klich
- Biostatistics Department, Hospices Civils de Lyon, Lyon France. CNRS, UMR 5558, University of Lyon, Biometrics and Evolutionary Biology Laboratory, Biostatistics-Health Team, 69100 Villeurbanne, France
| | - Sophie Trouillet-Assant
- Joint Research Unit Hospices Civils of Lyon-bioMérieux, Lyon Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France.
| |
Collapse
|
30
|
Pardi N, Krammer F. mRNA vaccines for infectious diseases - advances, challenges and opportunities. Nat Rev Drug Discov 2024; 23:838-861. [PMID: 39367276 DOI: 10.1038/s41573-024-01042-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2024] [Indexed: 10/06/2024]
Abstract
The concept of mRNA-based vaccines emerged more than three decades ago. Groundbreaking discoveries and technological advancements over the past 20 years have resolved the major roadblocks that initially delayed application of this new vaccine modality. The rapid development of nucleoside-modified COVID-19 mRNA vaccines demonstrated that this immunization platform is easy to develop, has an acceptable safety profile and can be produced at a large scale. The flexibility and ease of antigen design have enabled mRNA vaccines to enter development for a wide range of viruses as well as for various bacteria and parasites. However, gaps in our knowledge limit the development of next-generation mRNA vaccines with increased potency and safety. A deeper understanding of the mechanisms of action of mRNA vaccines, application of novel technologies enabling rational antigen design, and innovative vaccine delivery strategies and vaccination regimens will likely yield potent novel vaccines against a wide range of pathogens.
Collapse
Affiliation(s)
- Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
31
|
Cobey S. Vaccination against rapidly evolving pathogens and the entanglements of memory. Nat Immunol 2024; 25:2015-2023. [PMID: 39384979 DOI: 10.1038/s41590-024-01970-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/26/2024] [Indexed: 10/11/2024]
Abstract
Immune memory determines infection risk and responses to future infections and vaccinations over potentially decades of life. Despite its centrality, the dynamics of memory to antigenically variable pathogens remains poorly understood. This Review examines how past exposures shape B cell responses to vaccinations with influenza and SARS-CoV-2. An overriding feature of vaccinations with these pathogens is the recall of primary responses, often termed 'imprinting' or 'original antigenic sin'. These recalled responses can inhibit the generation of new responses unless some incompletely defined conditions are met. Depending on the context, immune memory can increase or decrease the total neutralizing antibody response to variant antigens, with apparent consequences for protection. These effects are easier to measure experimentally than epidemiologically, but there is evidence that both early and recent exposures influence vaccine effectiveness. A few immunological interactions between adaptive immune responses and antigens might explain the seemingly discrepant effects of memory. Overall, the complex observations point to a need for more quantitative approaches to integrate high-dimensional immune data from populations with diverse exposure histories. Such approaches could help identify optimal vaccination strategies against antigenically diverse pathogens.
Collapse
Affiliation(s)
- Sarah Cobey
- Department of Ecology and Evolution, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
32
|
Mucker EM, Freyn AW, Bixler SL, Cizmeci D, Atyeo C, Earl PL, Natarajan H, Santos G, Frey TR, Levin RH, Meni A, Arunkumar GA, Stadlbauer D, Jorquera PA, Bennett H, Johnson JC, Hardcastle K, Americo JL, Cotter CA, Koehler JW, Davis CI, Shamblin JD, Ostrowski K, Raymond JL, Ricks KM, Carfi A, Yu WH, Sullivan NJ, Moss B, Alter G, Hooper JW. Comparison of protection against mpox following mRNA or modified vaccinia Ankara vaccination in nonhuman primates. Cell 2024; 187:5540-5553.e10. [PMID: 39236707 DOI: 10.1016/j.cell.2024.08.043] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 08/09/2024] [Accepted: 08/21/2024] [Indexed: 09/07/2024]
Abstract
In 2022, mpox virus (MPXV) spread worldwide, causing 99,581 mpox cases in 121 countries. Modified vaccinia Ankara (MVA) vaccine use reduced disease in at-risk populations but failed to deliver complete protection. Lag in manufacturing and distribution of MVA resulted in additional MPXV spread, with 12,000 reported cases in 2023 and an additional outbreak in Central Africa of clade I virus. These outbreaks highlight the threat of zoonotic spillover by Orthopoxviruses. mRNA-1769, an mRNA-lipid nanoparticle (LNP) vaccine expressing MPXV surface proteins, was tested in a lethal MPXV primate model. Similar to MVA, mRNA-1769 conferred protection against challenge and further mitigated symptoms and disease duration. Antibody profiling revealed a collaborative role between neutralizing and Fc-functional extracellular virion (EV)-specific antibodies in viral restriction and ospinophagocytic and cytotoxic antibody functions in protection against lesions. mRNA-1769 enhanced viral control and disease attenuation compared with MVA, highlighting the potential for mRNA vaccines to mitigate future pandemic threats.
Collapse
Affiliation(s)
- Eric M Mucker
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | | | - Sandra L Bixler
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | | | | | - Patricia L Earl
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | - Jeffrey L Americo
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Catherine A Cotter
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jeff W Koehler
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Christopher I Davis
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Joshua D Shamblin
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Kristin Ostrowski
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Jo Lynne Raymond
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Keersten M Ricks
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | | | | | - Nancy J Sullivan
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, USA; Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA, USA; Department of Biology, Boston University, Boston, MA, USA
| | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Jay W Hooper
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA.
| |
Collapse
|
33
|
Kanokudom S, Chansaenroj J, Suntronwong N, Wongsrisang L, Aeemjinda R, Vichaiwattana P, Thatsanathorn T, Chantima W, Pakchotanon P, Duangchinda T, Sudhinaraset N, Honsawek S, Poovorawan Y. Safety and antibody responses of Omicron BA.4/5 bivalent booster vaccine among hybrid immunity with diverse vaccination histories: A cohort study. Vaccine X 2024; 20:100538. [PMID: 39211731 PMCID: PMC11359987 DOI: 10.1016/j.jvacx.2024.100538] [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: 01/02/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 09/04/2024] Open
Abstract
This cohort study, conducted between July and August 2023, evaluated the adverse events (AEs) and immune response to a bivalent mRNA-1273.222 (containing sequences of the original Wuhan-H1 strain and the Omicron BA.4/5 variant) booster vaccine in 122 participants. The study included individuals with diverse vaccination histories, and their responses were assessed based on anti-receptor binding domain (RBD) IgG levels and neutralizing antibodies against the wild-type, Omicron BA.5, and XBB.1.16 variants. Following administration of the BA.4/5 bivalent vaccine, AEs were generally mild to moderate and well-tolerated within a few days. There were no reports of vomiting and no serious AEs or death. The findings demonstrated robust immune responses, with significant increases in anti-RBD IgG levels, particularly in groups that had received 3 -6 doses before the booster dose. The BA.4/5 bivalent booster effectively induced neutralizing antibodies against the vaccine strains, providing robust neutralization, including the XBB.1.16 strain. The study also highlighted that individuals with hybrid immunity, especially those assumed infected with the BA.5 strain or who had been infected twice, showed higher levels of robust neutralizing activity against Omicron XBB.1.16. Overall, these results indicate that the BA.4/5 bivalent booster vaccines can induce potent and good antibody responses in emerging Omicron subvariants, supporting its efficacy as a booster in individuals with diverse vaccination histories.
Collapse
Affiliation(s)
- Sitthichai Kanokudom
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Osteoarthritis and Musculoskeleton, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok 10330, Thailand
| | - Jira Chansaenroj
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Nungruthai Suntronwong
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Lakkhana Wongsrisang
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Ratchadawan Aeemjinda
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Preeyaporn Vichaiwattana
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Thaksaporn Thatsanathorn
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Warangkana Chantima
- Division of Dengue Hemorrhagic Fever Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Pattarakul Pakchotanon
- Molecular Biology of Dengue and Flaviviruses Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| | - Thaneeya Duangchinda
- Molecular Biology of Dengue and Flaviviruses Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| | - Natthinee Sudhinaraset
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Sittisak Honsawek
- Center of Excellence in Osteoarthritis and Musculoskeleton, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok 10330, Thailand
| | - Yong Poovorawan
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Fellow of the Royal Society of Thailand (FRS [T]), The Royal Society of Thailand, Sanam Sueapa, Dusit, Bangkok 10300, Thailand
| |
Collapse
|
34
|
Nilles EJ, Roberts K, de St Aubin M, Mayfield H, Restrepo AC, Garnier S, Abdalla G, Etienne MC, Duke W, Dumas D, Jarolim P, Oasan T, Peña F, Lopez B, Cruz LDL, Sanchez IM, Murray K, Baldwin M, Skewes-Ramm R, Paulino CT, Lau CL, Kucharski A. Convergence of SARS-CoV-2 spike antibody levels to a population immune setpoint. EBioMedicine 2024; 108:105319. [PMID: 39232463 PMCID: PMC11404201 DOI: 10.1016/j.ebiom.2024.105319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 08/06/2024] [Accepted: 08/17/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND Individual immune responses to SARS-CoV-2 are well-studied, while the combined effect of these responses on population-level immune dynamics remains poorly understood. Given the key role of population immunity on pathogen transmission, delineation of the factors that drive population immune evolution has critical public health implications. METHODS We enrolled individuals 5 years and older selected using a multistage cluster survey approach in the Northwest and Southeast of the Dominican Republic. Paired blood samples were collected mid-pandemic (Aug 2021) and late pandemic (Nov 2022). We measured serum pan-immunoglobulin antibodies against the SARS-CoV-2 spike protein. Generalized Additive Models (GAMs) and random forest models were used to analyze the relationship between changes in antibody levels and various predictor variables. Principal component analysis and partial dependence plots further explored the relationships between predictors and antibody changes. FINDINGS We found a transformation in the distribution of antibody levels from an irregular to a normalized single peak Gaussian distribution that was driven by titre-dependent boosting. This led to the convergence of antibody levels around a common immune setpoint, irrespective of baseline titres and vaccination profile. INTERPRETATION Our results suggest that titre-dependent kinetics driven by widespread transmission direct the evolution of population immunity in a consistent manner. These findings have implications for targeted vaccination strategies and improved modeling of future transmission, providing a preliminary blueprint for understanding population immune dynamics that could guide public health and vaccine policy for SARS-CoV-2 and potentially other pathogens. FUNDING The study was primarily funded by the Centers for Disease Control and Prevention grant U01GH002238 (EN). Salary support was provided by Wellcome Trust grant 206250/Z/17/Z (AK) and the Australian National Health and Medical Research Council Investigator grant APP1158469 (CLL).
Collapse
Affiliation(s)
- Eric J Nilles
- Brigham and Womens Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Harvard Humanitarian Initiative, Cambridge, MA, USA.
| | - Kathryn Roberts
- Brigham and Womens Hospital, Boston, MA, USA; Harvard Humanitarian Initiative, Cambridge, MA, USA
| | - Michael de St Aubin
- Brigham and Womens Hospital, Boston, MA, USA; Harvard Humanitarian Initiative, Cambridge, MA, USA
| | | | | | - Salome Garnier
- Brigham and Womens Hospital, Boston, MA, USA; Harvard Humanitarian Initiative, Cambridge, MA, USA
| | | | | | - William Duke
- Pedro Henríquez Ureña National University, Santo Domingo, Dominican Republic
| | - Devan Dumas
- Brigham and Womens Hospital, Boston, MA, USA; Harvard Humanitarian Initiative, Cambridge, MA, USA
| | - Petr Jarolim
- Brigham and Womens Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | | | - Farah Peña
- Ministry of Health and Social Assistance, Santo Domingo, Dominican Republic
| | - Beatriz Lopez
- Centers for Disease Control and Prevention, Central America Regional Office, Guatemala City, Guatemala
| | - Lucia de la Cruz
- Ministry of Health and Social Assistance, Santo Domingo, Dominican Republic
| | | | - Kristy Murray
- Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Margaret Baldwin
- Brigham and Womens Hospital, Boston, MA, USA; Harvard Humanitarian Initiative, Cambridge, MA, USA
| | - Ronald Skewes-Ramm
- Ministry of Health and Social Assistance, Santo Domingo, Dominican Republic
| | | | | | - Adam Kucharski
- London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
35
|
Macchia I, La Sorsa V, Ciervo A, Ruspantini I, Negri D, Borghi M, De Angelis ML, Luciani F, Martina A, Taglieri S, Durastanti V, Altavista MC, Urbani F, Mancini F. T Cell Peptide Prediction, Immune Response, and Host-Pathogen Relationship in Vaccinated and Recovered from Mild COVID-19 Subjects. Biomolecules 2024; 14:1217. [PMID: 39456150 PMCID: PMC11505848 DOI: 10.3390/biom14101217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024] Open
Abstract
COVID-19 remains a significant threat, particularly to vulnerable populations. The emergence of new variants necessitates the development of treatments and vaccines that induce both humoral and cellular immunity. This study aimed to identify potentially immunogenic SARS-CoV-2 peptides and to explore the intricate host-pathogen interactions involving peripheral immune responses, memory profiles, and various demographic, clinical, and lifestyle factors. Using in silico and experimental methods, we identified several CD8-restricted SARS-CoV-2 peptides that are either poorly studied or have previously unreported immunogenicity: fifteen from the Spike and three each from non-structural proteins Nsp1-2-3-16. A Spike peptide, LA-9, demonstrated a 57% response rate in ELISpot assays using PBMCs from 14 HLA-A*02:01 positive, vaccinated, and mild-COVID-19 recovered subjects, indicating its potential for diagnostics, research, and multi-epitope vaccine platforms. We also found that younger individuals, with fewer vaccine doses and longer intervals since infection, showed lower anti-Spike (ELISA) and anti-Wuhan neutralizing antibodies (pseudovirus assay), higher naïve T cells, and lower central memory, effector memory, and CD4hiCD8low T cells (flow cytometry) compared to older subjects. In our cohort, a higher prevalence of Vδ2-γδ and DN T cells, and fewer naïve CD8 T cells, seemed to correlate with strong cellular and lower anti-NP antibody responses and to associate with Omicron infection, absence of confusional state, and habitual sporting activity.
Collapse
Affiliation(s)
- Iole Macchia
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Valentina La Sorsa
- Research Promotion and Coordination Service, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Alessandra Ciervo
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Irene Ruspantini
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Martina Borghi
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Maria Laura De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Francesca Luciani
- National Center for the Control and Evaluation of Medicines, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.L.); (A.M.)
| | - Antonio Martina
- National Center for the Control and Evaluation of Medicines, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.L.); (A.M.)
| | - Silvia Taglieri
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Valentina Durastanti
- Neurology Unit, San Filippo Neri Hospital, ASL RM1, 00135 Rome, Italy; (V.D.); (M.C.A.)
| | | | - Francesca Urbani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Fabiola Mancini
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| |
Collapse
|
36
|
O’Reilly S, Byrne J, Feeney ER, Mallon PWG, Gautier V. Navigating the Landscape of B Cell Mediated Immunity and Antibody Monitoring in SARS-CoV-2 Vaccine Efficacy: Tools, Strategies and Clinical Trial Insights. Vaccines (Basel) 2024; 12:1089. [PMID: 39460256 PMCID: PMC11511438 DOI: 10.3390/vaccines12101089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 10/28/2024] Open
Abstract
Correlates of Protection (CoP) are biomarkers above a defined threshold that can replace clinical outcomes as primary endpoints, predicting vaccine effectiveness to support the approval of new vaccines or follow up studies. In the context of COVID-19 vaccination, CoPs can help address challenges such as demonstrating vaccine effectiveness in special populations, against emerging SARS-CoV-2 variants or determining the durability of vaccine-elicited immunity. While anti-spike IgG titres and viral neutralising capacity have been characterised as CoPs for COVID-19 vaccination, the contribution of other components of the humoral immune response to immediate and long-term protective immunity is less well characterised. This review examines the evidence supporting the use of CoPs in COVID-19 clinical vaccine trials, and how they can be used to define a protective threshold of immunity. It also highlights alternative humoral immune biomarkers, including Fc effector function, mucosal immunity, and the generation of long-lived plasma and memory B cells and discuss how these can be applied to clinical studies and the tools available to study them.
Collapse
Affiliation(s)
- Sophie O’Reilly
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Joanne Byrne
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Eoin R. Feeney
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Department of Infectious Diseases, St Vincent’s University Hospital, Elm Park, Dublin 4, Ireland
| | - Patrick W. G. Mallon
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Department of Infectious Diseases, St Vincent’s University Hospital, Elm Park, Dublin 4, Ireland
| | - Virginie Gautier
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
37
|
Karismananda, Hasyim AA, Sakamoto A, Yamagata K, Zainal KH, Suparman DDN, Yustisia I, Hardjo M, Kadir S, Iyori M, Yoshida S, Yusuf Y. Long-Term Immunity against SARS-CoV-2 Wild-Type and Omicron XBB.1.5 in Indonesian Residents after Vaccination and Infection. Antibodies (Basel) 2024; 13:72. [PMID: 39311377 PMCID: PMC11417924 DOI: 10.3390/antib13030072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 08/12/2024] [Accepted: 08/21/2024] [Indexed: 09/26/2024] Open
Abstract
In the post-pandemic era, evaluating long-term immunity against COVID-19 has become increasingly critical, particularly in light of continuous SARS-CoV-2 mutations. This study aimed to assess the long-term humoral immune response in sera collected in Makassar. We measured anti-RBD IgG levels and neutralization capacity (NC) against both the Wild-Type (WT) Wuhan-Hu and Omicron XBB.1.5 variants across groups of COVID-19-vaccinated individuals with no booster (NB), single booster (SB), and double booster (DB). The mean durations since the last vaccination were 25.11 months, 19.24 months, and 16.9 months for the NB, SB, and DB group, respectively. Additionally, we evaluated the effect of breakthrough infection (BTI) history, with a mean duration since the last confirmed infection of 21.72 months. Our findings indicate fair long-term WT antibody (Ab) titers, with the DB group showing a significantly higher level than the other groups. Similarly, the DB group demonstrated the highest anti-Omicron XBB.1.5 Ab titer, yet it was insignificantly different from the other groups. Although the level of anti-WT Ab titers was moderate, we observed near-complete (96-97%) long-term neutralization against the WT pseudo-virus for all groups. There was a slight decrease in NC against Omicron XBB.1.5 compared to the WT among all groups, as DB group, SB group, and NB group showed 80.71 ± 3.9%, 74.29 ± 6.7%, and 67.2 ± 6.3% neutralization activity, respectively. A breakdown analysis based on infection and vaccine status showed that booster doses increase the NC against XBB.1.5, particularly in individuals without BTI. Individuals with BTI demonstrate a better NC compared to their counterpart uninfected individuals with the same number of booster doses. Our findings suggest that long-term immunity against SARS-CoV-2 persists and is effective against the mutant variant. Booster doses enhance the NC, especially among uninfected individuals.
Collapse
Affiliation(s)
- Karismananda
- Master Program of Biomedical Science, Graduate School of Hasanuddin University, Makassar 90245, Indonesia; (K.); (I.Y.); (M.H.); (S.K.)
| | - Ammar Abdurrahman Hasyim
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University, Kanazawa 920-1192, Japan; (A.A.H.); (K.Y.); (K.H.Z.); (S.Y.)
| | - Akihiko Sakamoto
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University, Kanazawa 920-1192, Japan; (A.A.H.); (K.Y.); (K.H.Z.); (S.Y.)
| | - Kyouhei Yamagata
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University, Kanazawa 920-1192, Japan; (A.A.H.); (K.Y.); (K.H.Z.); (S.Y.)
| | - Kartika Hardianti Zainal
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University, Kanazawa 920-1192, Japan; (A.A.H.); (K.Y.); (K.H.Z.); (S.Y.)
| | | | - Ika Yustisia
- Master Program of Biomedical Science, Graduate School of Hasanuddin University, Makassar 90245, Indonesia; (K.); (I.Y.); (M.H.); (S.K.)
- Department of Biochemistry, Faculty of Medicine, Hasanuddin University, Makassar 90245, Indonesia
| | - Marhaen Hardjo
- Master Program of Biomedical Science, Graduate School of Hasanuddin University, Makassar 90245, Indonesia; (K.); (I.Y.); (M.H.); (S.K.)
- Department of Biochemistry, Faculty of Medicine, Hasanuddin University, Makassar 90245, Indonesia
| | - Syahrijuita Kadir
- Master Program of Biomedical Science, Graduate School of Hasanuddin University, Makassar 90245, Indonesia; (K.); (I.Y.); (M.H.); (S.K.)
- Department of Biochemistry, Faculty of Medicine, Hasanuddin University, Makassar 90245, Indonesia
| | - Mitsuhiro Iyori
- Research Institute of Pharmaceutical Sciences, Musashino University, Nishitokyo 202-8585, Japan;
| | - Shigeto Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University, Kanazawa 920-1192, Japan; (A.A.H.); (K.Y.); (K.H.Z.); (S.Y.)
| | - Yenni Yusuf
- Department of Parasitology, Faculty of Medicine, Hasanuddin University, Makassar 90245, Indonesia
| |
Collapse
|
38
|
Jones CH, Hauguel T, Beitelshees M, Davitt M, Welch V, Lindert K, Allen P, True JM, Dolsten M. Deciphering immune responses: a comparative analysis of influenza vaccination platforms. Drug Discov Today 2024; 29:104125. [PMID: 39097221 DOI: 10.1016/j.drudis.2024.104125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/21/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
Influenza still poses a significant challenge due to its high mutation rates and the low effectiveness of traditional vaccines. At present, antibodies that neutralize the highly variable hemagglutinin antigen are a major driver of the observed variable protection. To decipher how influenza vaccines can be improved, an analysis of licensed vaccine platforms was conducted, contrasting the strengths and limitations of their different mechanisms of protection. Through this review, it is evident that these vaccines do not elicit the robust cellular immune response critical for protecting high-risk groups. Emerging platforms, such as RNA vaccines, that induce robust cellular responses that may be additive to the recognized mechanism of protection through hemagglutinin inhibition may overcome these constraints to provide broader, protective immunity. By combining both humoral and cellular responses, such platforms could help guide the future influenza vaccine development.
Collapse
Affiliation(s)
| | | | | | | | - Verna Welch
- Pfizer, Hudson Boulevard, New York, NY 10018, USA
| | | | - Pirada Allen
- Pfizer, Hudson Boulevard, New York, NY 10018, USA
| | - Jane M True
- Pfizer, Hudson Boulevard, New York, NY 10018, USA.
| | | |
Collapse
|
39
|
Tunheim G, Fossum E, Robertson AH, Rø GØI, Chopra A, Vaage JT, Vikse EL, Kran AMB, Magnus P, Trogstad L, Mjaaland S, Hungnes O, Lund-Johansen F. Characterization of the SARS-CoV-2 antibody landscape in Norway in the late summer of 2022: high seroprevalence in all age groups with patterns of primary Omicron infection in children and hybrid immunity in adults. BMC Infect Dis 2024; 24:841. [PMID: 39164637 PMCID: PMC11334563 DOI: 10.1186/s12879-024-09670-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 07/25/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND According to Norwegian registries, 91% of individuals ≥ 16 years had received ≥ 1 dose of COVID-19 vaccine by mid-July 2022, whereas less than 2% of children < 12 years were vaccinated. Confirmed COVID-19 was reported for 27% of the population, but relaxation of testing lead to substantial underreporting. We have characterized the humoral immunity to SARS-CoV-2 in Norway in the late summer of 2022 by estimating the seroprevalence and identifying antibody profiles based on reactivity to Wuhan or Omicron-like viruses in a nationwide cross-sectional collection of residual sera, and validated our findings using cohort sera. METHODS 1,914 anonymized convenience sera and 243 NorFlu-cohort sera previously collected from the Oslo-area with reported infection and vaccination status were analyzed for antibodies against spike, the receptor-binding domain (RBD) of the ancestral Wuhan strain and Omicron BA.2 RBD, and nucleocapsid (N). Samples were also tested for antibodies inhibiting RBD-ACE2 interaction. Neutralization assays were performed on subsets of residual sera against B.1, BA.2, XBB.1.5 and BQ.1.1. RESULTS The national seroprevalence estimate from vaccination and/or infection was 99.1% (95% CrI 97.0-100.0%) based on Wuhan (spike_W and RBD_W) and RBD_BA2 antibodies. Sera from children < 12 years had 2.2 times higher levels of antibodies against RBD_BA2 than RBD_W and their seroprevalence estimate showed a 14.4 percentage points increase when also including anti-RBD_BA2 antibodies compared to Wuhan-antibodies alone. 50.3% (95% CI 45.0-55.5%) of residual sera from children and 38.1% (95% CI 36.0-40.4%) of all residual sera were positive for anti-N-antibodies. By combining measurements of binding- and ACE2-RBD-interaction-inhibiting antibodies, reactivity profiles indicative of infection and vaccination history were identified and validated using cohort sera. Residual sera with a profile indicative of hybrid immunity were able to neutralize newer Omicron variants XBB.1.5 and BQ.1.1. CONCLUSIONS By late summer of 2022, most of the Norwegian population had antibodies to SARS-CoV-2, and almost all children had been infected. Antibody profiles indicated that children mostly had experienced a primary Omicron infection, while hybrid immunity was common among adults. The finding that sera displaying hybrid immunity could neutralize newer Omicron variants indicates that Wuhan-like priming of the immune response did not have a harmful imprinting effect and that infections induce cross-reacting antibodies against future variants.
Collapse
Affiliation(s)
- Gro Tunheim
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway.
| | - Even Fossum
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Anna Hayman Robertson
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | | | - Adity Chopra
- Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - John T Vaage
- Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Elisabeth Lea Vikse
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Anne-Marte Bakken Kran
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Per Magnus
- Center for Fertility and Health, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Lill Trogstad
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Siri Mjaaland
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Olav Hungnes
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | | |
Collapse
|
40
|
Ardyanto TD, Khariri K, Agus TP, Soebandrio A. Post COVID-19 vaccination binding and neutralizing antibody with or without previous infection: An 18-month longitudinal study in Indonesia. NARRA J 2024; 4:e1071. [PMID: 39280276 PMCID: PMC11394176 DOI: 10.52225/narra.v4i2.1071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 08/29/2024] [Indexed: 09/18/2024]
Abstract
Due to the persisting development of SARS-CoV-2 variants, studies on the kinetics, duration, and function of antibodies are essential for vaccine development and long-term immunity prediction. This longitudinal study examined post-vaccination antibody responses in people after receiving CoronaVac or ChAdOx1 vaccines with or without a history of SARS-CoV-2 infection. Conducted in Indonesia between August 2021 and May 2023, this study involved 121 participants divided into two groups based on the received vaccine types and monitored for 18 months post-second dose vaccination by assessing the binding antibody (BAb) level and neutralizing antibody (NAb) inhibition rate at six time points. The study also documented the participants' age, gender, and body mass index (BMI). Before the first dose vaccination, 85 (70.2%) participants were reactive BAb (defined by BAb level ≥50 AU/mL) indicating a history of infection. In the CoronaVac group, only 53.1% were reactive BAb. However, 100% of participants were positive NAb (defined by NAb inhibition rate ≥30%), which indicates a past history of infection with low initial or rapidly decreasing BAb levels. In the ChAdOx1 group, 81.9% of participants were reactive, while only 54.2% were positive NAb, suggesting a recent infection with a high BAb level but a relatively low NAb inhibition rate. During the 18 months post-second dose vaccination, the BAb levels fluctuated. However, 100% of participants were positive NAb. No significant difference in antibody response was documented among participants with or without infection history. Also, no significant impact was presented by the factors of sex, age, and BMI. The findings highlight the crucial of the vaccine in public health and how vaccination strategies could be optimized effectively during and after the post-pandemic.
Collapse
Affiliation(s)
- Tonang D Ardyanto
- Department of Clinical Pathology, Faculty of Medicine, Universitas Sebelas Maret, Surakarta, Indonesia
| | - Khariri Khariri
- Doctoral Program of Biomedical Science, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- National Research and Innovation Agency, Jakarta, Indonesia
| | - Telly P Agus
- National Research and Innovation Agency, Jakarta, Indonesia
| | - Amin Soebandrio
- Department of Clinical Microbiology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
41
|
Ricketson LJ, Doucette EJ, Alatorre I, Tarannum T, Gray J, Booth W, Tipples G, Charlton C, Kanji JN, Fonseca K, Kellner JD. Pediatric antibody responses to SARS-CoV-2 after infection and vaccination in Calgary, Canada. BMC Infect Dis 2024; 24:705. [PMID: 39026179 PMCID: PMC11256562 DOI: 10.1186/s12879-024-09615-3] [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: 04/03/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND There are few reports of longitudinal serologic responses in children following Sars-CoV-2 infection and vaccination. This study describes longitudinal SARS-CoV-2 antibody responses following infection, vaccination, or both (hybrid immunity) in a cohort of Canadian children. The objectives of our study were to compare antibody levels following SARS-CoV-2 infection, vaccination, and hybrid immunity and to examine antibody decline after final antigen exposure. METHODS The Alberta Childhood COVID-19 Cohort (AB3C) study was a prospective longitudinal cohort study conducted from July 2020 to September 2022 with repeat sampling across 5 visits. Children under 18 years of age were enrolled for serial measurement of antibody responses to SARS-CoV-2 virus vaccine and infection. RESULTS The final sample size was 919; participants were 50.5% female, 48.2% were > 12 years and 88.5% were white ethnicity. The median peak spike IgG level of those with only infection was not different from those with no vaccination or infection (233 AU/mL (IQR: 99-944 AU/mL) vs. 3 AU/mL (IQR: 1-5 AU/mL; P = 0.1765). Participants with infections after vaccination had higher IgG levels than those where infection preceded vaccination (median: 36,660 (IQR: 22,084 - 40,000 AU/mL) vs. 17,461 AU/mL (IQR: 10,617 - 33,212 AU/mL); P < 0.0001). In a linear mixed methods model, children with infection-only had low levels of antibody that stayed stable over the study duration without further antigen exposures. Those with infection after vaccination had the slowest rate of antibody decline over time at 4% (95%CI: 2-5%) per week, compared with children where infection preceded vaccine 7% (95%CI: 6-8%) per week. CONCLUSIONS Children with hybrid immunity conferred through vaccination (2 + doses) followed by a SARS-CoV-2 infection had the highest and longest lasting antibody levels, compared to children who had an infection followed by vaccination, vaccination-only, or infection-only. The longer-term clinical importance of these findings, related to prevention of repeated infections and severe outcomes and need for further vaccine doses, is not yet known.
Collapse
Affiliation(s)
- Leah J Ricketson
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Emily J Doucette
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Isabella Alatorre
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Tarannum Tarannum
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Joslyn Gray
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - William Booth
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Graham Tipples
- Public Health Laboratory, Alberta Precision Laboratories, Calgary, AB, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Carmen Charlton
- Public Health Laboratory, Alberta Precision Laboratories, Calgary, AB, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jamil N Kanji
- Public Health Laboratory, Alberta Precision Laboratories, Calgary, AB, Canada
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Division of Infectious Diseases, Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Microbiology, Immunology & Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - Kevin Fonseca
- Public Health Laboratory, Alberta Precision Laboratories, Calgary, AB, Canada
- Department of Microbiology, Immunology & Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - James D Kellner
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
42
|
Fantin RF, Clark JJ, Cohn H, Jaiswal D, Bozarth B, Civljak A, Rao V, Lobo I, Nardulli JR, Srivastava K, Yong J, Andreata-Santos R, Bushfield K, Lee ES, Singh G, PVI Study Group, Kleinstein SH, Krammer F, Simon V, Bajic G, Coelho CH. Dissecting human monoclonal antibody responses from mRNA- and protein-based XBB.1.5 COVID-19 monovalent vaccines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.15.602781. [PMID: 39071292 PMCID: PMC11275766 DOI: 10.1101/2024.07.15.602781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
The emergence of highly contagious and immune-evasive severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants has required reformulation of coronavirus disease 2019 (COVID-19) vaccines to target those new variants specifically. While previous infections and booster vaccinations can enhance variant neutralization, it is unclear whether the monovalent version, administered using either mRNA or protein-based vaccine platforms, can elicit de novo B-cell responses specific for Omicron XBB.1.5 variants. Here, we dissected the genetic antibody repertoire of 603 individual plasmablasts derived from five individuals who received a monovalent XBB.1.5 vaccination either with mRNA (Moderna or Pfizer/BioNtech) or adjuvanted protein (Novavax). From these sequences, we expressed 100 human monoclonal antibodies and determined binding, affinity and protective potential against several SARS-CoV-2 variants, including JN.1. We then select two vaccine-induced XBB.1.5 mAbs, M2 and M39. M2 mAb was a de novo, antibody, i.e., specific for XBB.1.5 but not ancestral SARS-CoV-2. M39 bound and neutralized both XBB.1.5 and JN.1 strains. Our high-resolution cryo-electron microscopy (EM) structures of M2 and M39 in complex with the XBB.1.5 spike glycoprotein defined the epitopes engaged and revealed the molecular determinants for the mAbs' specificity. These data show, at the molecular level, that monovalent, variant-specific vaccines can elicit functional antibodies, and shed light on potential functional and genetic differences of mAbs induced by vaccinations with different vaccine platforms.\.
Collapse
Affiliation(s)
- Raianna F. Fantin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jordan J. Clark
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hallie Cohn
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Deepika Jaiswal
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bailey Bozarth
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alesandro Civljak
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vishal Rao
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Igor Lobo
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica R. Nardulli
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Komal Srivastava
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jeremy Yong
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Andreata-Santos
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Retrovirology Laboratory, Department of Microbiology, Immunology and Parasitology, Paulista School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Kaitlyn Bushfield
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Edward S. Lee
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - PVI Study Group
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steven H. Kleinstein
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
- Program in Computational Biology and Biomedical Informatics, Yale University, New Haven, CT 06520, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| | - Viviana Simon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Goran Bajic
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Camila H. Coelho
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
43
|
Ganusov VV. Appropriate Sampling and Longer Follow-Up Are Required to Rigorously Evaluate Longevity of Humoral Memory After Vaccination. Immunohorizons 2024; 8:397-403. [PMID: 38864816 PMCID: PMC11220738 DOI: 10.4049/immunohorizons.2300057] [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: 07/18/2023] [Accepted: 04/25/2024] [Indexed: 06/13/2024] Open
Abstract
One of the goals of vaccination is to induce long-lived immunity against the infection and/or disease. Many studies have followed the generation of humoral immunity to SARS-CoV-2 after vaccination; however, such studies typically varied by the duration of the follow-up and the number of time points at which immune response measurements were done. How these parameters (the number of time points and the overall duration of the follow-up) impact estimates of immunity longevity remain largely unknown. Several studies, including one by Arunachalam et al. (2023. J. Clin. Invest. 133: e167955), evaluated the humoral immune response in individuals receiving either a third or fourth dose of mRNA COVID-19 vaccine; by measuring Ab levels at three time points (prior to vaccination and at 1 and 6 mo), Arunachalam et al. found similar half-life times for serum Abs in the two groups and thus suggested that additional boosting is unnecessary to prolong immunity to SARS-CoV-2. I demonstrate that measuring Ab levels at these three time points and only for 6 mo does not allow one to accurately evaluate the long-term half-life of vaccine-induced Abs. By using the data from a cohort of blood donors followed for several years, I show that after revaccination with vaccinia virus, vaccinia virus-specific Abs decay biphasically, and even the late decay rate exceeds the true slow loss rate of humoral memory observed years prior to the boosting. Mathematical models of Ab response kinetics, parameterized using preliminary data, should be used for power analysis to determine the most appropriate timing and duration of sampling to rigorously determine the duration of humoral immunity after vaccination.
Collapse
Affiliation(s)
- Vitaly V. Ganusov
- Address correspondence and reprint request to: Vitaly V. Ganusov, Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227-5302. E-mail address:
| |
Collapse
|
44
|
Korosec CS, Dick DW, Moyles IR, Watmough J. SARS-CoV-2 booster vaccine dose significantly extends humoral immune response half-life beyond the primary series. Sci Rep 2024; 14:8426. [PMID: 38637521 PMCID: PMC11026522 DOI: 10.1038/s41598-024-58811-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 04/03/2024] [Indexed: 04/20/2024] Open
Abstract
SARS-CoV-2 lipid nanoparticle mRNA vaccines continue to be administered as the predominant prophylactic measure to reduce COVID-19 disease pathogenesis. Quantifying the kinetics of the secondary immune response from subsequent doses beyond the primary series and understanding how dose-dependent immune waning kinetics vary as a function of age, sex, and various comorbidities remains an important question. We study anti-spike IgG waning kinetics in 152 individuals who received an mRNA-based primary series (first two doses) and a subset of 137 individuals who then received an mRNA-based booster dose. We find the booster dose elicits a 71-84% increase in the median Anti-S half life over that of the primary series. We find the Anti-S half life for both primary series and booster doses decreases with age. However, we stress that although chronological age continues to be a good proxy for vaccine-induced humoral waning, immunosenescence is likely not the mechanism, rather, more likely the mechanism is related to the presence of noncommunicable diseases, which also accumulate with age, that affect immune regulation. We are able to independently reproduce recent observations that those with pre-existing asthma exhibit a stronger primary series humoral response to vaccination than compared to those that do not, and further, we find this result is sustained for the booster dose. Finally, via a single-variate Kruskal-Wallis test we find no difference between male and female humoral decay kinetics, however, a multivariate approach utilizing Least Absolute Shrinkage and Selection Operator (LASSO) regression for feature selection reveals a statistically significant (p < 1 × 10 - 3 ), albeit small, bias in favour of longer-lasting humoral immunity amongst males.
Collapse
Affiliation(s)
- Chapin S Korosec
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada.
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada.
| | - David W Dick
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada.
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada.
| | - Iain R Moyles
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada
| | - James Watmough
- Department of Mathematics and Statistics, University of New Brunswick, 3 Bailey Dr, Fredericton, E3B 5A3, NB, Canada
| |
Collapse
|