1
|
Sulcebe G, Ylli A, Cenko F, Kurti-Prifti M, Shyti E, Dashi-Pasholli J, Lazri E, Seferi-Qendro I, Perry MJ. Dynamics of anti-SARS-CoV-2 antibodies in the Albanian population: Impact of infection- and vaccine-induced immunity during the COVID-19 pandemic. IJID REGIONS 2024; 13:100440. [PMID: 39386114 PMCID: PMC11462266 DOI: 10.1016/j.ijregi.2024.100440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 10/12/2024]
Abstract
Objectives Understanding immune response dynamics during the COVID-19 pandemic is crucial for optimizing future vaccine strategies. This study investigated the infection- and vaccine-induced SARS-CoV-2 antibody responses in the Albanian population from August 2021 to August 2022. Methods This used a cross-sectional approach, analyzing two independent, randomly selected population samples over 1 year. Participants' demographic, health, vaccination, and COVID-19 data were collected, with blood samples assessed via enzyme linked immunosorbent assay for immunoglobulin G class anti-spike and anti-nucleocapsid antibodies. Results By August 2022, all individuals receiving one vaccine dose achieved antibody levels comparable to those receiving two doses (median 7.71 index ratio [IR] vs 7.00 IR). In August 2021, those with previous COVID-19 infection receiving one vaccine dose showed median anti-spike immunoglobulin G levels of 7.22 IR compared with 4.84 IR in those without previous infection receiving two doses. However, individuals aged ≥61 years required two vaccine doses to achieve similar immune responses as younger individuals with one dose. Conclusions These findings underscore the importance of hybrid immunity, suggesting one vaccine dose may suffice for individuals with previous COVID-19 infection, whereas older adults require additional doses for optimal protection. This study provides insights into humoral immune response dynamics, which is crucial for refining COVID-19 vaccination strategies in middle-income countries with low vaccination coverage and high infection rates.
Collapse
Affiliation(s)
- Genc Sulcebe
- Research Center of Biotechnology and Genetics, Academy of Sciences of Albania, Tirana, Albania
- University of Medicine of Tirana, Tirana, Albania
| | - Alban Ylli
- University of Medicine of Tirana, Tirana, Albania
- Institute of Public Health, Tirana, Albania
| | - Fabian Cenko
- Catholic University “Our Lady of Good Counsel” Tirana, Tirana, Albania
| | - Margarita Kurti-Prifti
- Research Center of Biotechnology and Genetics, Academy of Sciences of Albania, Tirana, Albania
| | - Erkena Shyti
- Research Center of Biotechnology and Genetics, Academy of Sciences of Albania, Tirana, Albania
| | - Jonida Dashi-Pasholli
- Research Center of Biotechnology and Genetics, Academy of Sciences of Albania, Tirana, Albania
| | - Erina Lazri
- University of Medicine of Tirana, Tirana, Albania
| | | | | |
Collapse
|
2
|
Donadeu L, Gomez-Olles S, Casanova F, Torija A, Lopez-Meseguer M, Boada-Pérez M, Kervella D, Crespo E, Carrera-Muñoz C, Campos-Varela I, Castells L, Cortese MF, Esperalba J, Fernández-Naval C, Quintero J, Muñoz M, Agüero F, Gonzalez-Costello J, Lladó L, Favà A, Cañas L, del Mar de la Hoz-Caballero M, Meneghini M, Torres IB, Juvé M, Hafkamp FMJ, Vila M, Robles AG, Buzón MJ, Toapanta N, Zúñiga JM, Monforte V, Saez-Giménez B, Len O, Arcos IL, Miret E, Ariceta G, Pardo E, Martínez X, Moreso F, Bestard O. Role of SARS-CoV-2-specific memory B cells promoting immune protection after booster vaccination in solid organ transplantation. Front Immunol 2024; 15:1463769. [PMID: 39439787 PMCID: PMC11493670 DOI: 10.3389/fimmu.2024.1463769] [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: 07/12/2024] [Accepted: 09/09/2024] [Indexed: 10/25/2024] Open
Abstract
Introduction Solid organ transplant (SOT) recipients display weak seroconversion and neutralizing antibody (NAb) responses after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination and remain at risk of severe coronavirus disease 2019 (COVID-19). While B-cell memory is the hallmark of serological immunity, its role in driving successful vaccine responses and providing immune protection in SOT patients remains unclear. Methods We investigated the function and interplay of SARS-CoV-2-specific memory B cells (mBc), different cytokineproducing T cells, and cross-reactive NAb in driving seroconversion and protection against COVID-19 in two cohorts. First, we studied a large cohort of 148 SOT recipients and 32 immunocompetent individuals who underwent several vaccinations. Subsequently, we assessed 25 SOT patients participating in a randomized controlled trial to compare two different immunosuppressive strategies for allowing successful seroconversion and memory-cell responses after booster vaccination. Results We corroborate previous findings that B- and T-cell memory responses are weaker and more delayed in SOT patients than in immunocompetent (IC) individuals; however, within the SOT cohort, we found that these responses are relatively stronger and more robust in patients not receiving mycophenolate mofetil (MMF)-based therapies. Anti- spike IgG titers strongly correlated with RBD-specific IgG-producing mBc, with both displaying broad viral cross reactivity. Prebooster SARS-CoV-2-specific mBc and IL-2- producing T cells accurately predicted Nab seroconversion (AUC, 0.828) and protection against severe COVID-19. While switching unresponsive SOT patients from calcineurin inhibitors (CNI)/MMF to a low-exposure CNI/mTOR-i regimen favored wider SARS-CoV-2-specific immune responses after a fourth booster vaccination, preformed RBD-specific mBc predicted NAb seroconversion. Discussion Our study adds new insights into the pathobiology of immune memory and highlights the pivotal role of SARS-CoV-2-specific mBc in promoting immune protection inSOT patients.
Collapse
Affiliation(s)
- Laura Donadeu
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Susana Gomez-Olles
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Laboratory of Pneumology, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Franc Casanova
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alba Torija
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Manuel Lopez-Meseguer
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Laboratory of Pneumology, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Lung Transplant Unit, Pneumology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Meritxell Boada-Pérez
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Laboratory of Pneumology, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Delphine Kervella
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Elena Crespo
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Claudia Carrera-Muñoz
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Isabel Campos-Varela
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Liver Unit, Vall d'Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Lluís Castells
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Liver Unit, Vall d'Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maria F. Cortese
- Microbiology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Juliana Esperalba
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Microbiology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Candela Fernández-Naval
- Microbiology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jesús Quintero
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Pediatric Hepatology and Liver Transplant Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marina Muñoz
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Pediatric Nephrology, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Fernando Agüero
- Department of Preventive Medicine and Epidemiology, Bellvitge University Hospital, Barcelona, Spain
| | - José Gonzalez-Costello
- Advanced Heart Failure and Heart Transplant Unit, Department of Cardiology, Hospital Universitari de Bellvitge, BIOHEART-Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute (IDIBELL), Universitat de Barcelona, Ciber Cardiovascular (CIBERCV), Barcelona, Spain
| | - Laura Lladó
- Liver Transplant Unit, Bellvitge University Hospital, Barcelona, Spain
| | - Alexandre Favà
- Kidney Transplant Unit, Bellvitge University Hospital, Barcelona, Spain
| | - Laura Cañas
- Kidney Transplant Unit, Nephrology department, Germans Trias i Pujol Hospital, Badalona, Spain
| | - María del Mar de la Hoz-Caballero
- Equipo de Atención Primaria Sant Rafael, Servei d'Atenció Primària (SAP) Muntanya, Gerència Territorial de Barcelona Ciutat, Institut Català de la Salut, Barcelona, Spain
| | - Maria Meneghini
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Irina B. Torres
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mariona Juvé
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - FMJ Hafkamp
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marta Vila
- Microbiology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alba G. Robles
- Infectious Diseases Department, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maria José Buzón
- Infectious Diseases Department, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Nestor Toapanta
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - José Miguel Zúñiga
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Víctor Monforte
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Laboratory of Pneumology, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Lung Transplant Unit, Pneumology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Berta Saez-Giménez
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Laboratory of Pneumology, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Lung Transplant Unit, Pneumology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Oscar Len
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Infectious Diseases, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ibai Los Arcos
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Infectious Diseases, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Enric Miret
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Urology Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Gema Ariceta
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Pediatric Nephrology, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Emma Pardo
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Xavier Martínez
- Department of Preventive Medicine and Epidemiology, Vall d’Hebron Hospital Universitari, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Francesc Moreso
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Oriol Bestard
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
3
|
Smith CP, Hartman RM, Kugler AM, Little V, Baker OR, Fairlie TA, Fernandez RE, Hagen MB, Honie E, Laeyendecker O, Midgley CM, Parker D, Sandoval M, Takahashi S, Hammitt LL, Sutcliffe CG. The Trajectory of Antibody Responses One Year Following SARS-CoV-2 Infection among Indigenous Individuals in the Southwest United States. Viruses 2024; 16:1573. [PMID: 39459907 PMCID: PMC11512241 DOI: 10.3390/v16101573] [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: 08/09/2024] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
SARS-CoV-2 antibody kinetics based on immunologic history is not fully understood. We analyzed anti-spike and anti-nucleocapsid antibody responses following acute infection in a cohort of Indigenous persons. The models of peak concentrations and decay rates estimated that one year after infection, participants would serorevert for anti-nucleocapsid antibodies and remain seropositive for anti-spike antibodies. The peak anti-spike concentrations were higher for individuals vaccinated prior to infection, but the decay rates were similar across immunologic status groups. Children had significantly lower peak anti-spike concentrations than adults. This study affirms the importance of continued vaccination to maintain high levels of immunity in the face of waning immunity.
Collapse
Affiliation(s)
- Claire P. Smith
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Rachel M. Hartman
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Alexa M. Kugler
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Verlena Little
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Owen R. Baker
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Tarayn A. Fairlie
- Division of Coronaviruses and Other Respiratory Viruses, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Reinaldo E. Fernandez
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Melissa B. Hagen
- Division of Coronaviruses and Other Respiratory Viruses, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Elvira Honie
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Oliver Laeyendecker
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Claire M. Midgley
- Division of Coronaviruses and Other Respiratory Viruses, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Dennie Parker
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Marqia Sandoval
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Saki Takahashi
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Laura L. Hammitt
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Catherine G. Sutcliffe
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
4
|
Invernizzi A, Renzetti S, van Thriel C, Rechtman E, Patrono A, Ambrosi C, Mascaro L, Corbo D, Cagna G, Gasparotti R, Reichenberg A, Tang CY, Lucchini RG, Wright RO, Placidi D, Horton MK. COVID-19 related cognitive, structural and functional brain changes among Italian adolescents and young adults: a multimodal longitudinal case-control study. Transl Psychiatry 2024; 14:402. [PMID: 39358346 PMCID: PMC11447249 DOI: 10.1038/s41398-024-03108-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) has been associated with brain functional, structural, and cognitive changes that persist months after infection. Most studies of the neurologic outcomes related to COVID-19 focus on severe infection and aging populations. Here, we investigated the neural activities underlying COVID-19 related outcomes in a case-control study of mildly infected youth enrolled in a longitudinal study in Lombardy, Italy, a global hotspot of COVID-19. All participants (13 cases, 27 controls, mean age 24 years) completed resting-state functional (fMRI), structural MRI, cognitive assessments (CANTAB spatial working memory) at baseline (pre-COVID) and follow-up (post-COVID). Using graph theory eigenvector centrality (EC) and data-driven statistical methods, we examined differences in ECdelta (i.e., the difference in EC values pre- and post-COVID-19) and Volumetricdelta (i.e., the difference in cortical volume of cortical and subcortical areas pre- and post-COVID) between COVID-19 cases and controls. We found that ECdelta significantly between COVID-19 and healthy participants in five brain regions; right intracalcarine cortex, right lingual gyrus, left hippocampus, left amygdala, left frontal orbital cortex. The left hippocampus showed a significant decrease in Volumetricdelta between groups (p = 0.041). The reduced ECdelta in the left amygdala associated with COVID-19 status mediated the association between COVID-19 and disrupted spatial working memory. Our results show persistent structural, functional and cognitive brain changes in key brain areas associated with olfaction and cognition. These results may guide treatment efforts to assess the longevity, reversibility and impact of the observed brain and cognitive changes following COVID-19.
Collapse
Affiliation(s)
- Azzurra Invernizzi
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Stefano Renzetti
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Christoph van Thriel
- Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
| | - Elza Rechtman
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alessandra Patrono
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Claudia Ambrosi
- Department of Neuroscience, Neuroradiology Unit, ASST Cremona, Cremona, Italy
| | | | - Daniele Corbo
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Giuseppa Cagna
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Roberto Gasparotti
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Abraham Reichenberg
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cheuk Y Tang
- Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Roberto G Lucchini
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, USA
| | - Robert O Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Donatella Placidi
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Megan K Horton
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
5
|
Rappuoli R, Alter G, Pulendran B. Transforming vaccinology. Cell 2024; 187:5171-5194. [PMID: 39303685 DOI: 10.1016/j.cell.2024.07.021] [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: 01/24/2024] [Revised: 06/24/2024] [Accepted: 07/12/2024] [Indexed: 09/22/2024]
Abstract
The COVID-19 pandemic placed the field of vaccinology squarely at the center of global consciousness, emphasizing the vital role of vaccines as transformative public health tools. The impact of vaccines was recently acknowledged by the award of the 2023 Nobel Prize in Physiology or Medicine to Katalin Kariko and Drew Weissman for their seminal contributions to the development of mRNA vaccines. Here, we provide a historic perspective on the key innovations that led to the development of some 27 licensed vaccines over the past two centuries and recent advances that promise to transform vaccines in the future. Technological revolutions such as reverse vaccinology, synthetic biology, and structure-based design transformed decades of vaccine failures into successful vaccines against meningococcus B and respiratory syncytial virus (RSV). Likewise, the speed and flexibility of mRNA vaccines profoundly altered vaccine development, and the advancement of novel adjuvants promises to revolutionize our ability to tune immunity. Here, we highlight exciting new advances in the field of systems immunology that are transforming our mechanistic understanding of the human immune response to vaccines and how to predict and manipulate them. Additionally, we discuss major immunological challenges such as learning how to stimulate durable protective immune response in humans.
Collapse
Affiliation(s)
| | - Galit Alter
- Moderna Therapeutics, Cambridge, MA 02139, USA.
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA; Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
6
|
Mvula M, Mtonga F, Mandolo J, Jowati C, Kalirani A, Chigamba P, Lisimba E, Mitole N, Chibwana MG, Jambo KC. Longevity of hybrid immunity against SARS-CoV-2 in adults vaccinated with an adenovirus-based COVID-19 vaccine. BMC Infect Dis 2024; 24:959. [PMID: 39266969 PMCID: PMC11391831 DOI: 10.1186/s12879-024-09891-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/05/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Hybrid immunity provides better protection against COVID-19 than vaccination or prior natural infection alone. It induces high magnitude and broadly cross-reactive neutralising anti-Spike IgG antibodies. However, it is not clear how long these potent antibodies last, especially in the context of adenovirus-based COVID-19 vaccines. METHODS We conducted a longitudinal cohort study and enrolled 20 adults who had received an adenovirus-based COVID-19 vaccine before a laboratory-confirmed SARS-CoV-2 infection. We followed up the study participants for 390 days post the initial breakthrough infection. We assessed the longevity and cross-reactive breadth of serum antibodies against SARS-CoV-2 variants of concern (VOCs), including Omicron. RESULTS The binding anti-Spike IgG antibodies remained within the reported putative levels for at least 360 days and were cross-neutralising against Beta, Gamma, Delta, and Omicron. During the follow up period, a median of one SARS-CoV-2 re-infection event was observed across the cohort, but none resulted in severe COVID-19. Moreover, the re-exposure events were associated with augmented anti-Spike and anti-RBD IgG antibody titres. CONCLUSIONS This study confirms that hybrid immunity provides durable broadly cross-reactive antibody immunity against SARS-CoV-2 variants of concern for at least a year (360 days), and that it is further augment by SARS-CoV-2 re-exposure.
Collapse
Affiliation(s)
- Memory Mvula
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - Fatima Mtonga
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - Jonathan Mandolo
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
- Liverpool School of Tropical Medicine, Liverpool, UK
| | - Chisomo Jowati
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - Alice Kalirani
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | | | - Edwin Lisimba
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - Ndaona Mitole
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - Marah G Chibwana
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
- University of Oxford, Oxford, England, UK
| | - Kondwani C Jambo
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi.
- Liverpool School of Tropical Medicine, Liverpool, UK.
| |
Collapse
|
7
|
Yan X, Zhao X, Du Y, Wang H, Liu L, Wang Q, Liu J, Wei S. Dynamics of anti-SARS-CoV-2 IgG antibody responses following breakthrough infection and the predicted protective efficacy: A longitudinal community-based population study in China. Int J Infect Dis 2024; 145:107075. [PMID: 38697605 DOI: 10.1016/j.ijid.2024.107075] [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: 01/14/2024] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/05/2024] Open
Abstract
OBJECTIVES To assess the dynamics of the anti-SARS-CoV-2 IgG antibody levels and their efficacy against COVID-19. METHODS We conducted a longitudinal serological analysis of 852 breakthrough COVID-19 infections among the community-based population in Yichang, China. Anti-SARS-CoV-2 IgG levels were measured by chemiluminescence at approximately 3, 4, and 9 months after infection. A linear mixed model predicted IgG antibody decline over 18 months. The effectiveness of antibodies in preventing symptomatic and severe infections was determined using an existing meta-regression model. RESULTS IgG antibodies slowly declined after breakthrough infections. Initially high at around 3 months (339.44 AU/mL, IQR: 262.78-382.95 AU/mL), levels remained significant at 9 months (297.74 AU/mL, IQR: 213.22-360.62 AU/mL). The elderly (≥60 years) had lower antibody levels compared to the young (<20 years) (P < 0.001). The protective efficacy of antibodies against symptomatic and severe infections was lower in the elderly (≥60 years) (78.34% and 86.33%) compared to the young (<20 years) (96.56% and 98.75%) after 1 year. CONCLUSION The study indicated a slow decline in anti-SARS-CoV-2 IgG antibodies, maintaining considerable efficacy for over 1 year. However, lower levels in the elderly suggest reduced protective effects, underscoring the need for age-specific vaccination strategies.
Collapse
Affiliation(s)
- Xiaolong Yan
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Zhao
- Center for Disease Control and Prevention, Yichang, Hubei, China
| | - Yin Du
- Center for Disease Control and Prevention, Yichang, Hubei, China
| | - Hao Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianhua Liu
- Center for Disease Control and Prevention, Yichang, Hubei, China
| | - Sheng Wei
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
8
|
Bouros I, Hill EM, Keeling MJ, Moore S, Thompson RN. Prioritising older individuals for COVID-19 booster vaccination leads to optimal public health outcomes in a range of socio-economic settings. PLoS Comput Biol 2024; 20:e1012309. [PMID: 39116038 PMCID: PMC11309497 DOI: 10.1371/journal.pcbi.1012309] [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: 03/08/2024] [Accepted: 07/09/2024] [Indexed: 08/10/2024] Open
Abstract
The rapid development of vaccines against SARS-CoV-2 altered the course of the COVID-19 pandemic. In most countries, vaccinations were initially targeted at high-risk populations, including older individuals and healthcare workers. Now, despite substantial infection- and vaccine-induced immunity in host populations worldwide, waning immunity and the emergence of novel variants continue to cause significant waves of infection and disease. Policy makers must determine how to deploy booster vaccinations, particularly when constraints in vaccine supply, delivery and cost mean that booster vaccines cannot be administered to everyone. A key question is therefore whether older individuals should again be prioritised for vaccination, or whether alternative strategies (e.g. offering booster vaccines to the individuals who have most contacts with others and therefore drive infection) can instead offer indirect protection to older individuals. Here, we use mathematical modelling to address this question, considering SARS-CoV-2 transmission in a range of countries with different socio-economic backgrounds. We show that the population structures of different countries can have a pronounced effect on the impact of booster vaccination, even when identical booster vaccination targeting strategies are adopted. However, under the assumed transmission model, prioritising older individuals for booster vaccination consistently leads to the most favourable public health outcomes in every setting considered. This remains true for a range of assumptions about booster vaccine supply and timing, and for different assumed policy objectives of booster vaccination.
Collapse
Affiliation(s)
- Ioana Bouros
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - Edward M. Hill
- Mathematics Institute, University of Warwick, Coventry, United Kingdom
- Zeeman Institute for Systems Biology and Infectious Disease Epidemiology Research (SBIDER), University of Warwick, Coventry, United Kingdom
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Matt J. Keeling
- Mathematics Institute, University of Warwick, Coventry, United Kingdom
- Zeeman Institute for Systems Biology and Infectious Disease Epidemiology Research (SBIDER), University of Warwick, Coventry, United Kingdom
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Sam Moore
- Lancaster Medical School, Lancaster University, Lancaster, United Kingdom
| | - Robin N. Thompson
- Mathematical Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
9
|
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
|
10
|
Movsisyan M, Truzyan N, Kasparova I, Chopikyan A, Sawaqed R, Bedross A, Sukiasyan M, Dilbaryan K, Shariff S, Kantawala B, Hakobjanyan G, Petrosyan G, Hakobyan A, Yenkoyan K. Tracking the evolution of anti-SARS-CoV-2 antibodies and long-term humoral immunity within 2 years after COVID-19 infection. Sci Rep 2024; 14:13417. [PMID: 38862731 PMCID: PMC11167004 DOI: 10.1038/s41598-024-64414-9] [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: 02/15/2024] [Accepted: 06/08/2024] [Indexed: 06/13/2024] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that gave rise to COVID-19 infection produced a worldwide health crisis. The virus can cause a serious or even fatal disease. Comprehending the complex immunological responses triggered by SARS-CoV-2 infection is essential for identifying pivotal elements that shape the course of the disease and its enduring effects on immunity. The span and potency of antibody responses provide valuable perspicuity into the resilience of post-infection immunity. The analysis of existing literature reveals a diverse controversy, confining varying data about the persistence of particular antibodies as well as the multifaceted factors that impact their development and titer, Within this study we aimed to understand the dynamics of anti-SARS-CoV-2 antibodies against nucleocapsid (anti-SARS-CoV-2 (N)) and spike (anti-SARS-CoV-2 (N)) proteins in long-term immunity in convalescent patients, as well as the factors influencing the production and kinetics of those antibodies. We collected 6115 serum samples from 1611 convalescent patients at different post-infection intervals up to 21 months Study showed that in the fourth month, the anti-SARS-CoV-2 (N) exhibited their peak mean value, demonstrating a 79% increase compared to the initial month. Over the subsequent eight months, the peak value experienced a modest decline, maintaining a relatively elevated level by the end of study. Conversely, anti-SARS-CoV-2 (S) exhibited a consistent increase at each three-month interval over the 15-month period, culminating in a statistically significant peak mean value at the study's conclusion. Our findings demonstrate evidence of sustained seropositivity rates for both anti-SARS-CoV-2 (N) and (S), as well as distinct dynamics in the long-term antibody responses, with anti-SARS-CoV-2 (N) levels displaying remarkable persistence and anti-SARS-CoV-2 (S) antibodies exhibiting a progressive incline.
Collapse
Affiliation(s)
- Mariam Movsisyan
- Department of Allergology and Clinical Immunology, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
- Cobrain Center, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Nune Truzyan
- Cobrain Center, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Irina Kasparova
- Department of Histology, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Armine Chopikyan
- Cobrain Center, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
- Department of Public Health and Healthcare Organization, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Ra'ed Sawaqed
- General Medicine Faculty, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Alexandra Bedross
- General Medicine Faculty, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Meline Sukiasyan
- Department of Allergology and Clinical Immunology, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
- Cobrain Center, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Karen Dilbaryan
- Cobrain Center, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Sanobar Shariff
- General Medicine Faculty, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Burhan Kantawala
- Cobrain Center, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Gohar Hakobjanyan
- Cobrain Center, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
- Laboratory-Diagnostic Center of Heratsi Clinical Hospital, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Gayane Petrosyan
- Cobrain Center, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
- Laboratory-Diagnostic Center of Heratsi Clinical Hospital, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Armine Hakobyan
- Department of Allergology and Clinical Immunology, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Konstantin Yenkoyan
- Neuroscience Laboratory, Cobrain Center, Yerevan State Medical University Named After Mkhitar Heratsi, 0025, Yerevan, Armenia.
- Department of Biochemistry, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia.
| |
Collapse
|
11
|
Abril AG, Alejandre J, Mariscal A, Alserawan L, Rabella N, Roman E, Lopez-Contreras J, Navarro F, Serrano E, Nomdedeu JF, Vidal S. Titers of IgG and IgA against SARS-CoV-2 proteins and their association with symptoms in mild COVID-19 infection. Sci Rep 2024; 14:12725. [PMID: 38830902 PMCID: PMC11148197 DOI: 10.1038/s41598-024-59634-y] [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: 09/21/2022] [Accepted: 04/12/2024] [Indexed: 06/05/2024] Open
Abstract
Humoral immunity in COVID-19 includes antibodies (Abs) targeting spike (S) and nucleocapsid (N) SARS-CoV-2 proteins. Antibody levels are known to correlate with disease severity, but titers are poorly reported in mild or asymptomatic cases. Here, we analyzed the titers of IgA and IgG against SARS-CoV-2 proteins in samples from 200 unvaccinated Hospital Workers (HWs) with mild COVID-19 at two time points after infection. We analyzed the relationship between Ab titers and patient characteristics, clinical features, and evolution over time. Significant differences in IgG and IgA titers against N, S1 and S2 proteins were found when samples were segregated according to time T1 after infection, seroprevalence at T1, sex and age of HWs and symptoms at infection. We found that IgM + samples had higher titers of IgG against N antigen and IgA against S1 and S2 antigens than IgM - samples. There were significant correlations between anti-S1 and S2 Abs. Interestingly, IgM + patients with dyspnea had lower titers of IgG and IgA against N, S1 and S2 than those without dyspnea. Comparing T1 and T2, we found that IgA against N, S1 and S2 but only IgG against certain Ag decreased significantly. In conclusion, an association was established between Ab titers and the development of infection symptoms.
Collapse
Affiliation(s)
- Andrés G Abril
- Departament Biologia Cel·lular, Facultat de Medicina, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193, Bellaterra, Spain
- Althaia Xarxa Assistencial Universitària de Manresa, 08243, Manresa, Spain
- Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central (IRIS-CC), 08500, Vic, Spain
| | - Jose Alejandre
- Grup de Malalties Inflamatòries, IIB-Sant Pau, Institut Recerca Hospital de la Santa Creu i Sant Pau, 08041, Barcelona, Spain
| | - Anais Mariscal
- Servei d'Immunologia, Hospital de la Santa Creu i Sant Pau, 08041, Barcelona, Spain
| | - Leticia Alserawan
- Servei d'Immunologia, Hospital de la Santa Creu i Sant Pau, 08041, Barcelona, Spain
| | - Nuria Rabella
- Servei de Microbiologia, Hospital de la Santa Creu i Sant Pau, 08041, Barcelona, Spain
| | - Eva Roman
- Servei de Patologia Digestiva, Hospital de la Santa Creu i Sant Pau, 08041, Barcelona, Spain
| | - Joaquin Lopez-Contreras
- Servei de Malalties Infeccioses, Hospital de la Santa Creu i Sant Pau, 08041, Barcelona, Spain
| | - Ferran Navarro
- Servei de Microbiologia, Hospital de la Santa Creu i Sant Pau, 08041, Barcelona, Spain
| | | | - Josep F Nomdedeu
- Servei d'Hematologia, Hospital de la Santa Creu i Sant Pau, 08041, Barcelona, Spain
| | - Silvia Vidal
- Departament Biologia Cel·lular, Facultat de Medicina, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193, Bellaterra, Spain.
- Grup de Malalties Inflamatòries, IIB-Sant Pau, Institut Recerca Hospital de la Santa Creu i Sant Pau, 08041, Barcelona, Spain.
| |
Collapse
|
12
|
Høeg TB, Ladhani S, Prasad V. How methodological pitfalls have created widespread misunderstanding about long COVID. BMJ Evid Based Med 2024; 29:142-146. [PMID: 37748921 PMCID: PMC11137465 DOI: 10.1136/bmjebm-2023-112338] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/09/2023] [Indexed: 09/27/2023]
Affiliation(s)
- Tracy Beth Høeg
- Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
- Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Shamez Ladhani
- Public Health England, London, UK
- St George's University of London, London, UK
| | - Vinay Prasad
- Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
13
|
Montmaneix-Engels F, Dimeglio C, Staes L, Da Silva I, Porcheron M, Jougla I, Hérin F, Izopet J. Study of the cellular and humoral immune responses to SARS-CoV-2 vaccination. Heliyon 2024; 10:e29116. [PMID: 38601689 PMCID: PMC11004869 DOI: 10.1016/j.heliyon.2024.e29116] [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: 01/11/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 04/12/2024] Open
Abstract
Our understanding of cellular immunity in response to COVID-19 infection or vaccination is limited because of less commonly used techniques. We investigated both the cellular and humoral immune responses before and after the administration of a third dose of the SARS-CoV-2 vaccine among a group of healthcare workers. Cellular immunity was evaluated using the VIDAS interferon-gamma (IFNγ) RUO test, which enables automated measurement of IFNγ levels after stimulating peripheral blood lymphocytes. Booster doses significantly enhanced both cellular and humoral immunity. Concerning cellular response, the booster dose increased the percentage of positive IFNγ release assay (IGRA) results but no difference in IFNγ release was found. The cellular response was not associated with protection against SARS-CoV-2 infection. Interestingly, vaccinated and infected healthcare workers exhibited the highest levels of anti-spike and neutralizing antibodies. In conclusion, the IGRA is a simple method for measuring cellular immune responses after vaccination. However, its usefulness as a complement to the study of humoral responses is yet to be demonstrated in future research.
Collapse
Affiliation(s)
- Faustine Montmaneix-Engels
- INSERM UMR1291-CNRS UMR5051-University Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, 31300, Toulouse, France
- Toulouse III Paul Sabatier University, 31062, Toulouse, France
| | - Chloé Dimeglio
- INSERM UMR1291-CNRS UMR5051-University Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, 31300, Toulouse, France
- CHU Toulouse, Purpan Hospital, Virology Laboratory, 31300, Toulouse, France
| | - Laeticia Staes
- CHU Toulouse, Purpan Hospital, Virology Laboratory, 31300, Toulouse, France
| | - Isabelle Da Silva
- CHU Toulouse, Purpan Hospital, Virology Laboratory, 31300, Toulouse, France
| | - Marion Porcheron
- CHU Toulouse, Purpan Hospital, Virology Laboratory, 31300, Toulouse, France
| | - Isabelle Jougla
- Occupational Diseases Department, Toulouse University Hospital, 31000, Toulouse, France
| | - Fabrice Hérin
- Occupational Diseases Department, Toulouse University Hospital, 31000, Toulouse, France
- UMR1295, Joint Research Unit INSERM- University Toulouse III Paul Sabatier, Centre for Epidemiology and Research in Population Health Unit (CERPOP), 31000, Toulouse, France
| | - Jacques Izopet
- INSERM UMR1291-CNRS UMR5051-University Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, 31300, Toulouse, France
- Toulouse III Paul Sabatier University, 31062, Toulouse, France
- CHU Toulouse, Purpan Hospital, Virology Laboratory, 31300, Toulouse, France
| |
Collapse
|
14
|
Stave GM, Nabeel I, Durand-Moreau Q. Long COVID-ACOEM Guidance Statement. J Occup Environ Med 2024; 66:349-357. [PMID: 38588073 DOI: 10.1097/jom.0000000000003059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
ABSTRACT Persistent symptoms are common after acute COVID-19, often referred to as long COVID. Long COVID may affect the ability to perform activities of daily living, including work. Long COVID occurs more frequently in those with severe acute COVID-19. This guidance statement reviews the pathophysiology of severe acute COVID-19 and long COVID and provides pragmatic approaches to long COVID symptoms, syndromes, and conditions in the occupational setting. Disability laws and workers' compensation are also addressed.
Collapse
Affiliation(s)
- Gregg M Stave
- From the Division of Occupational and Environmental Medicine, Duke University, Durham, NC (G.M.S.); Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY (I.N.); and Division of Preventive Medicine, University of Alberta, Edmonton, Canada (Q.D.-M.)
| | | | | |
Collapse
|
15
|
Naffeti B, BenAribi W, Kebir A, Diarra M, Schoenhals M, Vigan-Womas I, Dellagi K, BenMiled S. Comparative reconstruction of SARS-CoV-2 transmission in three African countries using a mathematical model integrating immunity data. IJID REGIONS 2024; 10:100-107. [PMID: 38204927 PMCID: PMC10776948 DOI: 10.1016/j.ijregi.2023.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 01/12/2024]
Abstract
Objectives Africa has experienced fewer COVID-19 cases and deaths than other regions, with a contrasting epidemiological situation between countries, raising questions regarding the determinants of disease spread in Africa. Methods We built a susceptible-exposed-infected-recovered model including COVID-19 mortality data where recovery class is structured by specific immunization and modeled by a partial differential equation considering the opposed effects of immunity decline and immunization. This model was applied to Tunisia, Senegal, and Madagascar. Results Senegal and Tunisia experienced two epidemic phases. Initially, infections emerged in naive individuals and were limited by social distancing. Variants of concern (VOCs) were also introduced. The second phase was characterized by successive epidemic waves driven by new VOCs that escaped host immunity. Meanwhile, Madagascar demonstrated a different profile, characterized by longer intervals between epidemic waves, increasing the pool of susceptible individuals who had lost their protective immunity. The impact of vaccination on model parameters in Tunisia and Senegal was evaluated. Conclusions Loss of immunity and vaccination-induced immunity have played crucial role in controlling the African pandemic. SARS-CoV-2 has become endemic now and will continue to circulate in African populations. However, previous infections provide significant protection against severe diseases, thus providing a basis for future vaccination strategies.
Collapse
Affiliation(s)
- Bechir Naffeti
- University of Tunis el Manar, Institut Pasteur de Tunis, Bio-(Informatic, Mathematics and Statistic) BIMS-Lab LR09-IPT16, Tunis, Tunisia
| | - Walid BenAribi
- University of Tunis el Manar, Institut Pasteur de Tunis, Bio-(Informatic, Mathematics and Statistic) BIMS-Lab LR09-IPT16, Tunis, Tunisia
| | - Amira Kebir
- University of Tunis el Manar, Institut Pasteur de Tunis, Bio-(Informatic, Mathematics and Statistic) BIMS-Lab LR09-IPT16, Tunis, Tunisia
- University of Tunis, Institut préparatoire aux études d'ingénieurs de Tunis, Tunis, Tunisia
| | - Maryam Diarra
- Institut Pasteur de Dakar, Epidemiology, Clinical Research and Data Sciences Department, Dakar, Senegal
| | - Matthieu Schoenhals
- Institut Pasteur de Madagascar, Immunology of Infectious Diseases Unit, Antananarivo, Madagascar
| | - Inès Vigan-Womas
- Institut Pasteur de Dakar, Immuno-physiopathology and Infectious Diseases Department, Dakar, Senegal
| | | | - Slimane BenMiled
- University of Tunis el Manar, Institut Pasteur de Tunis, Bio-(Informatic, Mathematics and Statistic) BIMS-Lab LR09-IPT16, Tunis, Tunisia
| |
Collapse
|
16
|
Mendiboure V, Teiti I, Aubry M, Teissier A, Paoaafaite T, Vanhomwegen J, Manuguerra JC, Fontanet A, Cao-Lormeau VM, Madec Y. SARS-CoV-2 seroprevalence and associated factors of infection before and after the Delta wave in French Polynesia: a cross-sectional study. BMC Public Health 2024; 24:382. [PMID: 38317107 PMCID: PMC10840228 DOI: 10.1186/s12889-024-17869-4] [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: 08/22/2023] [Accepted: 01/24/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND French Polynesia (FP) comprises 75 inhabited islands scattered across five archipelagos. Between July and October 2021, the SARS-CoV-2 Delta variant triggered a much stronger second epidemic wave in FP than the original Wuhan strain, which was dominant from August 2020 to March 2021. Although previous seroprevalence surveys made it possible to determine the proportion of the population infected by SARS-CoV-2 on the two most populated islands (Tahiti and Moorea) after the first (20.6% in Tahiti and 9.4% in Moorea) and second (57.7% in Tahiti) epidemic waves, no data are available for more remote islands. We used blood samples and personal data collected before, during, and after the second wave from inhabitants of several islands within the five archipelagos to assess the prevalence of SARS-CoV-2 infections and identify associated factors. METHODS Blood samples and personal data were collected between April and December 2021 as part of the MATAEA study, a cross-sectional survey conducted on a random sample of the adult population representative of the five FP archipelagos and stratified by age and gender. IgG antibodies targeting the SARS-CoV-2 nucleocapsid (N) protein were detected using a recombinant antigen-based microsphere immunoassay. Factors associated with anti-SARS-CoV-2-N seropositivity were identified using logistic regression models. RESULTS Of 1,120 participants, 503 (44.9%) tested positive for anti-SARS-CoV-2-N antibodies, corresponding to a weighted prevalence of 56.8% for the FP population aged 18-69 years. The seroprevalence increased from 21.9% to 62.1% before and during/after the Delta wave. Of these infections, only 28.4% had been diagnosed by health professionals. The odds of being seropositive were lower in males, participants recruited before the Delta wave, those who had never been married, those with a diagnosed respiratory allergy, smokers, and those vaccinated against COVID-19. CONCLUSIONS Our results confirm the high impact of the Delta wave in FP. By the end of 2021, 56.8% of the FP population aged 18-69 years had been infected by SARS-CoV-2; the majority of these infections went undetected. Individuals with respiratory allergies were found to be less susceptible to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Vincent Mendiboure
- Emerging Diseases Epidemiology Unit, Institut Pasteur, Université Paris Cité, 75015, Paris, France
| | - Iotefa Teiti
- Laboratory of Research On Emerging Viral Diseases, Institut Louis Malardé, Papeete, Tahiti, French Polynesia
| | - Maite Aubry
- Laboratory of Research On Emerging Viral Diseases, Institut Louis Malardé, Papeete, Tahiti, French Polynesia
| | - Anita Teissier
- Laboratory of Research On Emerging Viral Diseases, Institut Louis Malardé, Papeete, Tahiti, French Polynesia
| | - Tuterarii Paoaafaite
- Laboratory of Research On Emerging Viral Diseases, Institut Louis Malardé, Papeete, Tahiti, French Polynesia
| | - Jessica Vanhomwegen
- Environment and Infectious Risks Unit, Laboratory for Urgent Response to Biological Threats (CIBU), Institut Pasteur, Université Paris Cité, 75015, Paris, France
| | - Jean-Claude Manuguerra
- Environment and Infectious Risks Unit, Laboratory for Urgent Response to Biological Threats (CIBU), Institut Pasteur, Université Paris Cité, 75015, Paris, France
| | - Arnaud Fontanet
- Emerging Diseases Epidemiology Unit, Institut Pasteur, Université Paris Cité, 75015, Paris, France
- Conservatoire National Des Arts Et Métiers, Unité PACRI, 75003, Paris, France
| | - Van-Mai Cao-Lormeau
- Laboratory of Research On Emerging Viral Diseases, Institut Louis Malardé, Papeete, Tahiti, French Polynesia
| | - Yoann Madec
- Emerging Diseases Epidemiology Unit, Institut Pasteur, Université Paris Cité, 75015, Paris, France.
| |
Collapse
|
17
|
Appelman B, Charlton BT, Goulding RP, Kerkhoff TJ, Breedveld EA, Noort W, Offringa C, Bloemers FW, van Weeghel M, Schomakers BV, Coelho P, Posthuma JJ, Aronica E, Joost Wiersinga W, van Vugt M, Wüst RCI. Muscle abnormalities worsen after post-exertional malaise in long COVID. Nat Commun 2024; 15:17. [PMID: 38177128 PMCID: PMC10766651 DOI: 10.1038/s41467-023-44432-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 12/13/2023] [Indexed: 01/06/2024] Open
Abstract
A subgroup of patients infected with SARS-CoV-2 remain symptomatic over three months after infection. A distinctive symptom of patients with long COVID is post-exertional malaise, which is associated with a worsening of fatigue- and pain-related symptoms after acute mental or physical exercise, but its underlying pathophysiology is unclear. With this longitudinal case-control study (NCT05225688), we provide new insights into the pathophysiology of post-exertional malaise in patients with long COVID. We show that skeletal muscle structure is associated with a lower exercise capacity in patients, and local and systemic metabolic disturbances, severe exercise-induced myopathy and tissue infiltration of amyloid-containing deposits in skeletal muscles of patients with long COVID worsen after induction of post-exertional malaise. This study highlights novel pathways that help to understand the pathophysiology of post-exertional malaise in patients suffering from long COVID and other post-infectious diseases.
Collapse
Affiliation(s)
- Brent Appelman
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Meibergdreef 9, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Braeden T Charlton
- Department of Human Movement Sciences, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Richie P Goulding
- Department of Human Movement Sciences, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Tom J Kerkhoff
- Department of Human Movement Sciences, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Movement Sciences, Amsterdam, the Netherlands
- Department of Physiology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Ellen A Breedveld
- Department of Human Movement Sciences, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Wendy Noort
- Department of Human Movement Sciences, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Carla Offringa
- Department of Human Movement Sciences, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Frank W Bloemers
- Amsterdam Movement Sciences, Amsterdam, the Netherlands
- Department of Trauma Surgery, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Core Facility Metabolomics, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Bauke V Schomakers
- Laboratory Genetic Metabolic Diseases, Core Facility Metabolomics, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Pedro Coelho
- Serviço de Neurologia, Departamento de Neurociências e Saúde Mental, Hospital de Santa Maria, CHULN, Lisbon, Portugal
- Faculdade de Medicina, Centro de Estudos Egas Moniz, University of Lisbon, Lisbon, Portugal
- Department of (Neuro)pathology, Amsterdam Neuroscience, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Jelle J Posthuma
- Department of Trauma Surgery, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
- Flevoziekenhuis, Division of Surgery, Hospitaalweg 1, Almere, the Netherlands
| | - Eleonora Aronica
- Department of (Neuro)pathology, Amsterdam Neuroscience, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - W Joost Wiersinga
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Meibergdreef 9, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
- Division of Infectious Diseases, Department of Internal Medicine, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Michèle van Vugt
- Amsterdam Institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands.
- Division of Infectious Diseases, Tropical Medicine, Department of Medicine, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands.
| | - Rob C I Wüst
- Department of Human Movement Sciences, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
- Amsterdam Movement Sciences, Amsterdam, the Netherlands.
| |
Collapse
|
18
|
de Oliveira MI, Aciole MR, Neves PAF, Silva VPOE, Silva MPOE, de Lorena VMB, de Araújo PSR. A stronger antibody response in increased disease severity of SARS-CoV-2. BMC Infect Dis 2024; 24:17. [PMID: 38166763 PMCID: PMC10762998 DOI: 10.1186/s12879-023-08923-4] [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: 04/23/2023] [Accepted: 12/17/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND An assessment of the factors that interfere with serum levels and the persistence of anti-SARs-CoV-2 IgG antibodies is essential in order to estimate the risk of reinfection and to plan vaccination. We analyzed the impact of the severity of coronavirus disease 2019 (COVID-19) and the clinical and biological factors regarding the persistence of SARs-CoV-2 anti-spike protein (IgG-S) antibodies at 12 months. METHODS This was an observational, longitudinal study with individuals who had recovered from COVID-19 between August 2020 and June 2021. Peripheral blood samples were collected from volunteers who were hospitalized (SERIOUS COVID-19) and those who required no hospitalization (COVID-19 LIGHT). Samples were grouped according to days after symptom onset: up to 90, between 91 and 180, ≥ 180 days after symptom onset. A semiquantitative test for IgG anti-spike protein S1(IgG-S1) was used. RESULTS We analyzed 238 individuals who had recovered from COVID-19, of whom 87 had been hospitalized and 151 had not. They provided 148 and 220 samples, respectively. Among those hospitalized, males (65.5%), volunteers aged over 60 years (41.1%), comorbidities such as arterial hypertension (67.8%) and diabetes mellitus (37.9%) were most frequent. We observed higher median serum IgG-S1 titers among those who had recovered from COVID-19 and had been hospitalized, at all collection time intervals (p < 0.001). We observed a weak correlation of increasing age with humoral IgG-S1 response (Spearman correlation = 0.298). There was a greater probability of IgG-S1 antibody persistence over time among samples from hospitalized individuals compared to samples from non-hospitalized participants (p = 0.001). CONCLUSION This study has revealed higher titers and a higher probability of the persistence of IgG-S1 in severe cases after SARs-CoV-2 primary infection in unvaccinated recovered patients. Thus, in this study, the severe clinical presentation of COVID-19 was the main factor influencing serum levels and the persistence of IgG-S1 antibodies in COVID-19.
Collapse
Affiliation(s)
- Marta Iglis de Oliveira
- Department of Tropical Medicine, Medical Sciences Center, Federal University of Pernambuco, Recife, Pernambuco, Brazil
- Department of Infectious Diseases, Hospital das Clínicas, Brazilian Company of Hospital Services (EBSERH), Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Melayne Rocha Aciole
- Department of Tropical Medicine, Medical Sciences Center, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Patrícia Areias Feitosa Neves
- Department of Tropical Medicine, Medical Sciences Center, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | | | | | - Virginia Maria Barros de Lorena
- Department of Tropical Medicine, Medical Sciences Center, Federal University of Pernambuco, Recife, Pernambuco, Brazil
- Department of Immunology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Fiocruz-PE, Recife, Pernambuco, Brazil
| | - Paulo Sérgio Ramos de Araújo
- Department of Tropical Medicine, Medical Sciences Center, Federal University of Pernambuco, Recife, Pernambuco, Brazil.
- Department of Parasitology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Fiocruz-PE, Recife, Pernambuco, Brazil.
- Department of Infectious Diseases, Hospital das Clínicas, Brazilian Company of Hospital Services (EBSERH), Federal University of Pernambuco, Recife, Pernambuco, Brazil.
| |
Collapse
|
19
|
Mead MN, Seneff S, Wolfinger R, Rose J, Denhaerynck K, Kirsch S, McCullough PA. COVID-19 mRNA Vaccines: Lessons Learned from the Registrational Trials and Global Vaccination Campaign. Cureus 2024; 16:e52876. [PMID: 38274635 PMCID: PMC10810638 DOI: 10.7759/cureus.52876] [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] [Accepted: 01/24/2024] [Indexed: 01/27/2024] Open
Abstract
Our understanding of COVID-19 vaccinations and their impact on health and mortality has evolved substantially since the first vaccine rollouts. Published reports from the original randomized phase 3 trials concluded that the COVID-19 mRNA vaccines could greatly reduce COVID-19 symptoms. In the interim, problems with the methods, execution, and reporting of these pivotal trials have emerged. Re-analysis of the Pfizer trial data identified statistically significant increases in serious adverse events (SAEs) in the vaccine group. Numerous SAEs were identified following the Emergency Use Authorization (EUA), including death, cancer, cardiac events, and various autoimmune, hematological, reproductive, and neurological disorders. Furthermore, these products never underwent adequate safety and toxicological testing in accordance with previously established scientific standards. Among the other major topics addressed in this narrative review are the published analyses of serious harms to humans, quality control issues and process-related impurities, mechanisms underlying adverse events (AEs), the immunologic basis for vaccine inefficacy, and concerning mortality trends based on the registrational trial data. The risk-benefit imbalance substantiated by the evidence to date contraindicates further booster injections and suggests that, at a minimum, the mRNA injections should be removed from the childhood immunization program until proper safety and toxicological studies are conducted. Federal agency approval of the COVID-19 mRNA vaccines on a blanket-coverage population-wide basis had no support from an honest assessment of all relevant registrational data and commensurate consideration of risks versus benefits. Given the extensive, well-documented SAEs and unacceptably high harm-to-reward ratio, we urge governments to endorse a global moratorium on the modified mRNA products until all relevant questions pertaining to causality, residual DNA, and aberrant protein production are answered.
Collapse
Affiliation(s)
- M Nathaniel Mead
- Biology and Nutritional Epidemiology, Independent Research, Copper Hill, USA
| | - Stephanie Seneff
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, USA
| | - Russ Wolfinger
- Biostatistics and Epidemiology, Independent Research, Research Triangle Park, USA
| | - Jessica Rose
- Immunology and Public Health Research, Independent Research, Ottawa, CAN
| | - Kris Denhaerynck
- Epidemiology and Biostatistics, Independent Research, Basel, CHE
| | - Steve Kirsch
- Data Science, Independent Research, Los Angeles, USA
| | - Peter A McCullough
- Cardiology, Epidemiology, and Public Health, McCullough Foundation, Dallas, USA
- Cardiology, Epidemiology, and Public Health, Truth for Health Foundation, Tucson, USA
| |
Collapse
|
20
|
Duminuco A, Nardo A, Orofino A, Giunta G, Conticello C, Del Fabro V, Chiarenza A, Parisi MS, Figuera A, Leotta S, Milone G, Cupri A, Cambria D, Di Raimondo F, Romano A, Palumbo GA. Efficacy and safety of tixagevimab-cilgavimab versus SARS-CoV-2 breakthrough infection in the hematological conditions. Cancer 2024; 130:41-50. [PMID: 37658645 DOI: 10.1002/cncr.35005] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/20/2023] [Accepted: 07/20/2023] [Indexed: 09/03/2023]
Abstract
BACKGROUND Managing SARS-CoV-2 infection in frail and immunosuppressed patients still represents an open challenge, but, starting from the phase 3 PROVENT study, prophylaxis with tixagevimab-cilgavimab has improved the approach in this category of patients, guaranteeing a better outcome and inferior mortality. Real-life data in a heterogeneous cohort are few. METHODS The aim of this study is to evaluate the benefit of prophylaxis with tixagevimab-cilgavimab in a cohort of 202 patients affected by different hematological diseases (lymphoproliferative, myeloproliferative, autoimmune, patients recently receiving a bone marrow transplant), active (with ongoing treatment), or in watch-and-wait strategy, followed in our center, during a median follow-up of 249 (45-325) days. RESULTS An incidence of 44 breakthrough infections (21.8%) is reported, with no treatment-related adverse effects. Age ≥70 years, ongoing treatment (above all with monoclonal antibodies), baseline lymphoproliferative disorders, and prior virus exposure are identified as risk factors related to subsequent infection (p < 0.05). Moreover, the incidence is higher in low/nonresponse to prior vaccination (p = .002). Patients treated with tixagevimab-cilgavimab had a mild course of the infection and a reduction of the duration compared with preprophylaxis infection (11 vs. 15 days, p < .001). The concurrent treatment with anti-CD20 monoclonal antibodies and B-non-Hodgkin lymphoma still confers a higher duration of infection despite prophylaxis. No deaths attributable to the infection occurred. CONCLUSION Prophylaxis treatment seems to be a valid and safe strategy, although not preventing breakthrough infection, but the severe complications associated with the infection and the possible delays in administering lifesaving therapies from long positivity.
Collapse
Affiliation(s)
- Andrea Duminuco
- Division of Hematology and BMT Unit, A.O.U. Policlinico G.Rodolico-San Marco, Catania, Italy
| | - Antonella Nardo
- Division of Hematology and BMT Unit, A.O.U. Policlinico G.Rodolico-San Marco, Catania, Italy
| | - Alessandra Orofino
- Division of Hematology and BMT Unit, A.O.U. Policlinico G.Rodolico-San Marco, Catania, Italy
| | - Giuliana Giunta
- Division of Hematology and BMT Unit, A.O.U. Policlinico G.Rodolico-San Marco, Catania, Italy
| | - Concetta Conticello
- Division of Hematology and BMT Unit, A.O.U. Policlinico G.Rodolico-San Marco, Catania, Italy
| | - Vittorio Del Fabro
- Division of Hematology and BMT Unit, A.O.U. Policlinico G.Rodolico-San Marco, Catania, Italy
| | - Annalisa Chiarenza
- Division of Hematology and BMT Unit, A.O.U. Policlinico G.Rodolico-San Marco, Catania, Italy
| | - Marina S Parisi
- Division of Hematology and BMT Unit, A.O.U. Policlinico G.Rodolico-San Marco, Catania, Italy
| | - Amalia Figuera
- Division of Hematology and BMT Unit, A.O.U. Policlinico G.Rodolico-San Marco, Catania, Italy
| | - Salvatore Leotta
- Division of Hematology and BMT Unit, A.O.U. Policlinico G.Rodolico-San Marco, Catania, Italy
| | - Giuseppe Milone
- Division of Hematology and BMT Unit, A.O.U. Policlinico G.Rodolico-San Marco, Catania, Italy
| | - Alessandra Cupri
- Division of Hematology and BMT Unit, A.O.U. Policlinico G.Rodolico-San Marco, Catania, Italy
| | - Daniela Cambria
- Division of Hematology and BMT Unit, A.O.U. Policlinico G.Rodolico-San Marco, Catania, Italy
| | - Francesco Di Raimondo
- Division of Hematology and BMT Unit, A.O.U. Policlinico G.Rodolico-San Marco, Catania, Italy
- Dipartimento di Specialità Medico-Chirurgiche, CHIRMED, Sezione di Ematologia University of Catania, Catania, Italy
| | - Alessandra Romano
- Division of Hematology and BMT Unit, A.O.U. Policlinico G.Rodolico-San Marco, Catania, Italy
- Dipartimento di Specialità Medico-Chirurgiche, CHIRMED, Sezione di Ematologia University of Catania, Catania, Italy
| | - Giuseppe A Palumbo
- Division of Hematology and BMT Unit, A.O.U. Policlinico G.Rodolico-San Marco, Catania, Italy
- Department of Scienze Mediche Chirurgiche e Tecnologie Avanzate G.F. Ingrassia, University of Catania, Catania, Italy
| |
Collapse
|
21
|
Erice A, Prieto L, Caballero C. Long-Term Analyses of SARS-CoV-2 Humoral and T Cell Responses and Breakthrough SARS-CoV-2 Infections after Two Doses of BNT162b2 Followed by mRNA-1273 and Bivalent Omicron-Adapted BNT162b2 Vaccines: A Prospective Study over 2 Years in Non-Immunocompromised Individuals. Vaccines (Basel) 2023; 11:1835. [PMID: 38140239 PMCID: PMC10748336 DOI: 10.3390/vaccines11121835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/28/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Long-term analyses of the immune response following SARS-CoV-2 mRNA vaccines are essential to determining its characteristics and providing the basis for vaccination strategies. We conducted a prospective study in a cohort of 268 healthy adults followed for >2 years after two doses of BNT162b2. Antibodies targeting the receptor-binding domain of the S1 subunit of the spike of SARS-CoV-2 (anti-RBD) were measured at eight time points; T cell response was analyzed using an interferon-γ release assay. A total of 248 (93%) subjects received mRNA-1273 on month 9; 93 (35%) received the bivalent Omicron-adapted BNT162b2 vaccine between months 19 and 26. Breakthrough infections occurred in 215 (80%) participants, with frequencies unaffected by the additional vaccines. Anti-RBD declined over the initial 9 months, increased after mRNA-1273, and declined gradually thereafter. In 50 (17%) previously infected subjects, anti-RBD levels were significantly higher up to month 9 (p < 0.05) but subsequently declined below those of uninfected individuals. Anti-RBD titers protective against SARS-CoV-2 could not be defined. Most subjects developed a positive T cell response that remained after 26 months. Waning of protection against SARS-CoV-2 infection occurred over time, resulting in non-severe breakthrough infections in most participants. The evolution of anti-RBD suggests modulation of the immune response through immune imprinting.
Collapse
Affiliation(s)
- Alejo Erice
- Department of Internal Medicine, Hospital Asepeyo, 28823 Coslada, Spain
- Unidad de Apoyo a la Investigación, Facultad de Medicina, Universidad Francisco de Vitoria, 28223 Pozuelo de Alarcón, Spain; (L.P.); (C.C.)
| | - Lola Prieto
- Unidad de Apoyo a la Investigación, Facultad de Medicina, Universidad Francisco de Vitoria, 28223 Pozuelo de Alarcón, Spain; (L.P.); (C.C.)
| | - Cristina Caballero
- Unidad de Apoyo a la Investigación, Facultad de Medicina, Universidad Francisco de Vitoria, 28223 Pozuelo de Alarcón, Spain; (L.P.); (C.C.)
- Clinical Diagnostic Laboratory, Hospital Asepeyo, 28823 Coslada, Spain
| |
Collapse
|
22
|
Tong MZW, Sng JDJ, Carney M, Cooper L, Brown S, Lineburg KE, Chew KY, Collins N, Ignacio K, Airey M, Burr L, Joyce BA, Jayasinghe D, McMillan CLD, Muller DA, Adhikari A, Gallo LA, Dorey ES, Barrett HL, Gras S, Smith C, Good‐Jacobson K, Short KR. Elevated BMI reduces the humoral response to SARS-CoV-2 infection. Clin Transl Immunology 2023; 12:e1476. [PMID: 38050635 PMCID: PMC10693902 DOI: 10.1002/cti2.1476] [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: 09/17/2023] [Revised: 11/05/2023] [Accepted: 11/09/2023] [Indexed: 12/06/2023] Open
Abstract
Objective Class III obesity (body mass index [BMI] ≥ 40 kg m-2) significantly impairs the immune response to SARS-CoV-2 vaccination. However, the effect of an elevated BMI (≥ 25 kg m-2) on humoral immunity to SARS-CoV-2 infection and COVID-19 vaccination remains unclear. Methods We collected blood samples from people who recovered from SARS-CoV-2 infection approximately 3 and 13 months of post-infection (noting that these individuals were not exposed to SARS-CoV-2 or vaccinated in the interim). We also collected blood samples from people approximately 5 months of post-second dose COVID-19 vaccination (the majority of whom did not have a prior SARS-CoV-2 infection). We measured their humoral responses to SARS-CoV-2, grouping individuals based on a BMI greater or less than 25 kg m-2. Results Here, we show that an increased BMI (≥ 25 kg m-2), when accounting for age and sex differences, is associated with reduced antibody responses after SARS-CoV-2 infection. At 3 months of post-infection, an elevated BMI was associated with reduced antibody titres. At 13 months of post-infection, an elevated BMI was associated with reduced antibody avidity and a reduced percentage of spike-positive B cells. In contrast, no significant association was noted between a BMI ≥ 25 kg m-2 and humoral immunity to SARS-CoV-2 at 5 months of post-secondary vaccination. Conclusions Taken together, these data showed that elevated BMI is associated with an impaired humoral immune response to SARS-CoV-2 infection. The impairment of infection-induced immunity in individuals with a BMI ≥ 25 kg m-2 suggests an added impetus for vaccination rather than relying on infection-induced immunity.
Collapse
Affiliation(s)
- Marcus ZW Tong
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Julian DJ Sng
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Meagan Carney
- School of Mathematics and PhysicsThe University of QueenslandSt LuciaQLDAustralia
| | - Lucy Cooper
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVICAustralia
- Immunity Program, Biomedicine Discovery InstituteMonash UniversityClaytonVICAustralia
| | - Samuel Brown
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Katie E Lineburg
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Infection and Inflammation ProgramQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
| | - Keng Yih Chew
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Neve Collins
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Kirsten Ignacio
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Megan Airey
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Lucy Burr
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Infection and Inflammation ProgramQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
- Department of Respiratory MedicineMater HealthBrisbaneQLDAustralia
| | - Briony A Joyce
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Dhilshan Jayasinghe
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVICAustralia
- Department of Biochemistry and ChemistryLa Trobe Institute for Molecular Science, La Trobe UniversityBundooraVICAustralia
| | - Christopher LD McMillan
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
- Australian Infectious Diseases Research CentreThe University of QueenslandSt LuciaQLDAustralia
| | - David A Muller
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
- Australian Infectious Diseases Research CentreThe University of QueenslandSt LuciaQLDAustralia
| | - Anurag Adhikari
- Department of Biochemistry and ChemistryLa Trobe Institute for Molecular Science, La Trobe UniversityBundooraVICAustralia
| | - Linda A Gallo
- School of HealthUniversity of the Sunshine CoastPetrieQLDAustralia
| | - Emily S Dorey
- Mater ResearchThe University of QueenslandSouth BrisbaneQLDAustralia
| | - Helen L Barrett
- Mater ResearchThe University of QueenslandSouth BrisbaneQLDAustralia
- University of New South Wales MedicineKensingtonNSWAustralia
- Obstetric MedicineRoyal Hospital for WomenRandwickNSWAustralia
| | - Stephanie Gras
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVICAustralia
- Department of Biochemistry and ChemistryLa Trobe Institute for Molecular Science, La Trobe UniversityBundooraVICAustralia
| | - Corey Smith
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Infection and Inflammation ProgramQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
| | - Kim Good‐Jacobson
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVICAustralia
- Immunity Program, Biomedicine Discovery InstituteMonash UniversityClaytonVICAustralia
| | - Kirsty R Short
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
- Australian Infectious Diseases Research CentreThe University of QueenslandSt LuciaQLDAustralia
| |
Collapse
|
23
|
Taylor KM, Ricks KM, Kuehnert PA, Eick-Cost AA, Scheckelhoff MR, Wiesen AR, Clements TL, Hu Z, Zak SE, Olschner SP, Herbert AS, Bazaco SL, Creppage KE, Fan MT, Sanchez JL. Seroprevalence as an Indicator of Undercounting of COVID-19 Cases in a Large Well-Described Cohort. AJPM FOCUS 2023; 2:100141. [PMID: 37885754 PMCID: PMC10598697 DOI: 10.1016/j.focus.2023.100141] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Introduction Reported confirmed cases represent a small portion of overall true cases for many infectious diseases. The undercounting of true cases can be considerable when a significant portion of infected individuals are asymptomatic or minimally symptomatic, as is the case with COVID-19. Seroprevalence studies are an efficient way to assess the extent to which true cases are undercounted during a large-scale outbreak and can inform efforts to improve case identification and reporting. Methods A longitudinal seroprevalence study of active duty U.S. military members was conducted from May 2020 through June 2021. A random selection of service member serum samples submitted to the Department of Defense Serum Repository was analyzed for the presence of antibodies reactive to SARS-CoV-2. The monthly seroprevalence rates were compared with those of cumulative confirmed cases reported during the study period. Results Seroprevalence was 2.3% in May 2020 and increased to 74.0% by June 2021. The estimated true case count based on seroprevalence was 9.3 times greater than monthly reported cases at the beginning of the study period and fell to 1.7 by the end of the study. Conclusions In our sample, confirmed case counts significantly underestimated true cases of COVID-19. The increased availability of testing over the study period and enhanced efforts to detect asymptomatic and minimally symptomatic cases likely contributed to the fall in the seroprevalence to reported case ratio.
Collapse
Affiliation(s)
- Kevin M. Taylor
- Armed Forces Health Surveillance Division, Defense Health Agency, Silver Spring, Maryland
- Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Keersten M. Ricks
- United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland
| | - Paul A. Kuehnert
- United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland
| | - Angelia A. Eick-Cost
- Armed Forces Health Surveillance Division, Defense Health Agency, Silver Spring, Maryland
| | - Mark R. Scheckelhoff
- Armed Forces Health Surveillance Division, Defense Health Agency, Silver Spring, Maryland
| | - Andrew R. Wiesen
- Health Readiness Policy and Oversight, Office of the Assistant Secretary of Defense for Health Affairs, Washington, District of Columbia
| | - Tamara L. Clements
- United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland
| | - Zheng Hu
- Armed Forces Health Surveillance Division, Defense Health Agency, Silver Spring, Maryland
| | - Samantha E. Zak
- United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland
| | - Scott P. Olschner
- United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland
| | - Andrew S. Herbert
- United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland
| | - Sara L. Bazaco
- Armed Forces Health Surveillance Division, Defense Health Agency, Silver Spring, Maryland
| | - Kathleen E. Creppage
- Armed Forces Health Surveillance Division, Defense Health Agency, Silver Spring, Maryland
| | - Michael T. Fan
- Armed Forces Health Surveillance Division, Defense Health Agency, Silver Spring, Maryland
| | - Jose L. Sanchez
- Armed Forces Health Surveillance Division, Defense Health Agency, Silver Spring, Maryland
| |
Collapse
|
24
|
Xu M, O’Brien MP, Hooper AT, Forleo-Neto E, Isa F, Hou P, Chan KC, Cohen MS, Marovich MA, Hamilton JD, Hirshberg B, Herman GA, Musser BJ. Nasopharyngeal Viral Load Is the Major Driver of Incident Antibody Immune Response to SARS-CoV-2 Infection. Open Forum Infect Dis 2023; 10:ofad598. [PMID: 38111750 PMCID: PMC10727195 DOI: 10.1093/ofid/ofad598] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/29/2023] [Indexed: 12/20/2023] Open
Abstract
Background Virologic determinants of seroconversion to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection were defined in a post hoc analysis of prospectively studied vaccine- and infection-naïve individuals at high risk for coronavirus disease 2019 (COVID-19). Methods This phase 3 COVID-19 prevention trial (NCT04452318) with casirivimab and imdevimab was conducted in July 2020-February 2021, before widespread vaccine availability. Placebo-treated participants who were uninfected (SARS-CoV-2 quantitative reverse transcription polymerase chain reaction [RT-qPCR] negative) and seronegative were assessed weekly for 28 days (efficacy assessment period [EAP]) for COVID-19 symptoms and SARS-CoV-2 infection by RT-qPCR of nasopharyngeal swab samples and for serostatus by antinucleocapsid immunoglobulin (Ig) G. Regression-based modeling, including causal mediation analysis, estimated the effects of viral load on seroconversion. Results Of 157/1069 (14.7%) uninfected and seronegative (for antispike IgG, antispike IgA, and antinucleocapsid IgG) participants who became infected during the EAP, 105 (65%) seroconverted. The mean (SD) maximum viral load of seroconverters was 7.23 (1.68) log10 copies/mL vs 4.8 (2.2) log10 copies/mL in those who remained seronegative; viral loads of ∼6.0 log10 copies/mL better predicted seroconversion. The mean of the maximum viral load was 7.11 log10 copies/mL in symptomatic participants vs 5.58 log10 copies/mL in asymptomatic participants. The mean duration of detectable viral load was longer in seroconverted vs seronegative participants: 3.24 vs 1.63 weeks. Conclusions Maximum SARS-CoV-2 viral load is a major driver of seroconversion and symptomatic COVID-19, with high viral loads (∼6.0 log10 copies/mL) better predicting seroconversion. Serology underestimates infection rates, incidence, and prevalence of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Meng Xu
- Regeneron Pharmaceuticals, Inc., Tarrytown, NewYork, USA
| | | | | | | | - Flonza Isa
- Regeneron Pharmaceuticals, Inc., Tarrytown, NewYork, USA
| | - Peijie Hou
- Regeneron Pharmaceuticals, Inc., Tarrytown, NewYork, USA
| | - Kuo-Chen Chan
- Regeneron Pharmaceuticals, Inc., Tarrytown, NewYork, USA
| | - Myron S Cohen
- University of North Carolina Chapel Hill School of Medicine, Institute for Global Health and Infectious Diseases, Chapel Hill, North Carolina, USA
| | - Mary A Marovich
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | | | - Boaz Hirshberg
- Regeneron Pharmaceuticals, Inc., Tarrytown, NewYork, USA
| | - Gary A Herman
- Regeneron Pharmaceuticals, Inc., Tarrytown, NewYork, USA
| | - Bret J Musser
- Regeneron Pharmaceuticals, Inc., Tarrytown, NewYork, USA
| |
Collapse
|
25
|
Ismail NF, Rahman AE, Kulkarni D, Zhu F, Wang X, del Carmen Morales G, Srivastava A, Allen KE, Spinardi J, Kyaw MH, Nair H. Incidence and outcome of SARS-CoV-2 reinfection in the pre-Omicron era: A global systematic review and meta-analysis. J Glob Health 2023; 13:06051. [PMID: 37994839 PMCID: PMC10667793 DOI: 10.7189/jogh.13.06051] [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] [Indexed: 11/24/2023] Open
Abstract
Background With the emergence of new variants and sub-lineages of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), reinfections can significantly impact herd immunity, vaccination policies, and decisions on other public health measures. We conducted a systematic review and meta-analysis to synthesise the global evidence on SARS-CoV-2 reinfections in the pre-Omicron era. Methods We searched five global databases (MEDLINE, Embase, CINAHL Plus, Global Health, WHO COVID-19) on 12 May 2022 and 28 July 2023 and three Chinese databases (CNKI, Wanfang, CQvip) on 16 October 2022 for articles reporting incidence and outcomes of SARS-CoV-2 reinfection before the period of Omicron (B.1.1.529) predominance. We assessed risk of bias using Joanna Briggs Institute critical appraisal tools and conducted meta-analyses with random effects models to estimate the proportion of SARS-CoV-2 reinfection among initially infected cases and hospitalisation and mortality proportions among reinfected ones. Results We identified 7593 studies and extracted data from 64 included ones representing 21 countries. The proportion of SARS-CoV-2 reinfection was 1.16% (95% confidence interval (CI) = 1.01-1.33) based on 11 639 247 initially infected cases, with ≥45 days between the two infections. Healthcare providers (2.28%; 95% CI = 1.37-3.40) had a significantly higher risk of reinfection than the general population (1.00%; 95% CI = 0.81-1.20), while young adults aged 18 to 35 years (1.01%; 95% CI = 0.8-1.25) had a higher reinfection burden than other age groups (children <18 years old: 0.57%; 95% CI = 0.39-0.79, older adults aged 36-65 years old: 0.53%; 95% CI = 0.41-0.65, elderly >65 years old: 0.37%; 95% CI = 0.15-0.66). Among the reinfected cases, 8.12% (95% CI = 5.30-11.39) were hospitalised, 1.31% (95% CI = 0.29-2.83) were admitted to the intensive care unit, and 0.71% (95% CI = 0.02-2.01) died. Conclusions Our data suggest a relatively low risk of SARS-CoV-2 reinfection in the pre-Omicron era, but the risk of hospitalisation was relatively high among the reinfected cases. Considering the possibility of underdiagnosis, the reinfection burden may be underestimated. Registration PROSPERO: CRD42023449712.
Collapse
Affiliation(s)
- Nabihah Farhana Ismail
- Centre for Global Health, University of Edinburgh, Edinburgh, United Kingdom
- Communicable Disease Control Unit, Public Health Department, Johor State, Malaysia
| | - Ahmed Ehsanur Rahman
- Centre for Global Health, University of Edinburgh, Edinburgh, United Kingdom
- International Centre for Diarrhoeal Diseases Research, Bangladesh
| | - Durga Kulkarni
- Centre for Global Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Fuyu Zhu
- School of Public Health, Nanjing Medical University, Jiangsu, China
| | - Xin Wang
- Centre for Global Health, University of Edinburgh, Edinburgh, United Kingdom
- School of Public Health, Nanjing Medical University, Jiangsu, China
| | | | - Amit Srivastava
- Pfizer, Vaccines, Emerging Markets
- Orbital Therapeutics, United States of America
| | | | | | | | - Harish Nair
- Centre for Global Health, University of Edinburgh, Edinburgh, United Kingdom
- School of Public Health, Nanjing Medical University, Jiangsu, China
- MRC/Wits Rural Public Health and Health Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
26
|
De-Simone SG, Napoleão-Pêgo P, Lechuga GC, Carvalho JPRS, Monteiro ME, Morel CM, Provance DW. Mapping IgA Epitope and Cross-Reactivity between Severe Acute Respiratory Syndrome-Associated Coronavirus 2 and DENV. Vaccines (Basel) 2023; 11:1749. [PMID: 38140154 PMCID: PMC10747746 DOI: 10.3390/vaccines11121749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/12/2023] [Accepted: 11/16/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND The newly introduced COVID-19 vaccines have reduced disease severity and hospitalizations. However, they do not significantly prevent infection or transmission. In the same context, measuring IgM and IgG antibody levels is important, but it does not provide information about the status of the mucosal immune response. This article describes a comprehensive mapping of IgA epitopes of the S protein, its cross-reactivity, and the development of an ELISA-peptide assay. METHODS IgA epitope mapping was conducted using SPOT synthesis and sera from RT-qPCR COVID-19-positive patients. Specific and cross-reacting epitopes were identified, and an evolutionary analysis from the early Wuhan strain to the Omicron variant was performed using bioinformatics tools and a microarray of peptides. The selected epitopes were chemically synthesized and evaluated using ELISA-IgA. RESULTS A total of 40 IgA epitopes were identified with 23 in S1 and 17 in the S2 subunit. Among these, at least 23 epitopes showed cross-reactivity with DENV and other organisms and 24 showed cross-reactivity with other associated coronaviruses. Three MAP4 polypeptides were validated by ELISA, demonstrating a sensitivity of 90-99.96% and a specificity of 100%. Among the six IgA-RBD epitopes, only the SC/18 epitope of the Omicron variants (BA.2 and BA.2.12.1) presented a single IgA epitope. CONCLUSIONS This research unveiled the IgA epitome of the S protein and identified many epitopes that exhibit cross-reactivity with DENV and other coronaviruses. The S protein of variants from Wuhan to Omicron retains many conserved IgA epitopes except for one epitope (#SCov/18). The cross-reactivity with DENV suggests limitations in using the whole S protein or the S1/S2/RBD segment for IgA serological diagnostic tests for COVID-19. The expression of these identified specific epitopes as diagnostic biomarkers could facilitate monitoring mucosal immunity to COVID-19, potentially leading to more accurate diagnoses and alternative mucosal vaccines.
Collapse
Affiliation(s)
- Salvatore G. De-Simone
- Center for Technological Development in Health (CDTS)/National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (P.N.-P.); (G.C.L.); (J.P.R.S.C.); (M.E.M.); (C.M.M.); (D.W.P.J.)
- Epidemiology and Molecular Systematics Laboratory (LEMS), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil
- Program of Post-Graduation on Science and Biotechnology, Department of Molecular and Cellular Biology, Biology Institute, Federal Fluminense University, Niterói 22040-036, RJ, Brazil
- Program of Post-Graduation on Parasitic Biology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil
| | - Paloma Napoleão-Pêgo
- Center for Technological Development in Health (CDTS)/National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (P.N.-P.); (G.C.L.); (J.P.R.S.C.); (M.E.M.); (C.M.M.); (D.W.P.J.)
- Epidemiology and Molecular Systematics Laboratory (LEMS), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil
| | - Guilherme C. Lechuga
- Center for Technological Development in Health (CDTS)/National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (P.N.-P.); (G.C.L.); (J.P.R.S.C.); (M.E.M.); (C.M.M.); (D.W.P.J.)
- Epidemiology and Molecular Systematics Laboratory (LEMS), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil
| | - João P. R. S. Carvalho
- Center for Technological Development in Health (CDTS)/National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (P.N.-P.); (G.C.L.); (J.P.R.S.C.); (M.E.M.); (C.M.M.); (D.W.P.J.)
- Program of Post-Graduation on Science and Biotechnology, Department of Molecular and Cellular Biology, Biology Institute, Federal Fluminense University, Niterói 22040-036, RJ, Brazil
| | - Maria E. Monteiro
- Center for Technological Development in Health (CDTS)/National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (P.N.-P.); (G.C.L.); (J.P.R.S.C.); (M.E.M.); (C.M.M.); (D.W.P.J.)
- Program of Post-Graduation on Parasitic Biology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil
| | - Carlos M. Morel
- Center for Technological Development in Health (CDTS)/National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (P.N.-P.); (G.C.L.); (J.P.R.S.C.); (M.E.M.); (C.M.M.); (D.W.P.J.)
| | - David W. Provance
- Center for Technological Development in Health (CDTS)/National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (P.N.-P.); (G.C.L.); (J.P.R.S.C.); (M.E.M.); (C.M.M.); (D.W.P.J.)
- Epidemiology and Molecular Systematics Laboratory (LEMS), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil
| |
Collapse
|
27
|
Llorente F, Pérez-Ramírez E, Pérez-Olmeda M, Dafouz-Bustos D, Fernández-Pinero J, Martínez-Cortés M, Jiménez-Clavero MÁ. The Detection of SARS-CoV-2 Antibodies in an Exposed Human Population Is Biased by the Immunoassay Used: Implications in Serosurveillance. Pathogens 2023; 12:1360. [PMID: 38003824 PMCID: PMC10675702 DOI: 10.3390/pathogens12111360] [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: 09/07/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
The presence of SARS-CoV-2 antibodies was examined over 7 months in a population of essential service workers exposed during the first epidemic wave in Madrid (Spain). Results obtained with different serological assays were compared. Firstly, serum samples obtained in April 2020 were analyzed using eleven SARS-CoV-2 antibody detection methods, including seven ELISAs, two CLIAs and two LFAs. While all of the ELISA tests and the Roche eCLIA method showed good performance, it was poorer for the Abbott CLIA and LFA tests. Sera from 115 workers with serologically positive results in April were collected 2 and 7 months after the first sampling and were analyzed using five of the tests previously assessed. The results showed that while some ELISA tests consistently detected the presence of anti-SARS-CoV-2 antibodies even 7 months after first detection, other methods, such as the Abbott CLIA test, showed an important reduction in sensitivity for these mature antibodies. The sensitivity increased after establishing new cut-off values, calculated taking into account both recent and old infections, suggesting that an adjustment of assay parameters may improve the detection of individuals exposed to the infection.
Collapse
Affiliation(s)
- Francisco Llorente
- Centro de Investigación en Sanidad Animal (CISA-INIA), CSIC, Valdeolmos, 28130 Madrid, Spain (M.Á.J.-C.)
| | - Elisa Pérez-Ramírez
- Centro de Investigación en Sanidad Animal (CISA-INIA), CSIC, Valdeolmos, 28130 Madrid, Spain (M.Á.J.-C.)
| | - Mayte Pérez-Olmeda
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), 28029 Madrid, Spain
| | - Desirée Dafouz-Bustos
- Centro de Investigación en Sanidad Animal (CISA-INIA), CSIC, Valdeolmos, 28130 Madrid, Spain (M.Á.J.-C.)
| | - Jovita Fernández-Pinero
- Centro de Investigación en Sanidad Animal (CISA-INIA), CSIC, Valdeolmos, 28130 Madrid, Spain (M.Á.J.-C.)
| | | | - Miguel Ángel Jiménez-Clavero
- Centro de Investigación en Sanidad Animal (CISA-INIA), CSIC, Valdeolmos, 28130 Madrid, Spain (M.Á.J.-C.)
- Centro de Investigación Biomédica en Red de Epidemiologia y Salud Pública (CIBERESP), 28029 Madrid, Spain
| |
Collapse
|
28
|
Pathakumari B, Marty PK, Shah M, Van Keulen VP, Erskine CL, Block MS, Arias-Sanchez P, Escalante P, Peikert T. Convalescent Adaptive Immunity Is Highly Heterogenous after SARS-CoV-2 Infection. J Clin Med 2023; 12:7136. [PMID: 38002748 PMCID: PMC10672050 DOI: 10.3390/jcm12227136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/03/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
The optimal detection strategies for effective convalescent immunity after SARS-CoV-2 infection and vaccination remain unclear. The objective of this study was to characterize convalescent immunity targeting the SARS-CoV-2 spike protein using a multiparametric approach. At the beginning of the pandemic, we recruited 30 unvaccinated convalescent donors who had previously been infected with COVID-19 and 7 unexposed asymptomatic controls. Peripheral blood mononuclear cells (PBMCs) were obtained from leukapheresis cones. The humoral immune response was assessed by measuring serum anti-SARS-CoV-2 spike S1 subunit IgG via semiquantitative ELISA, and T-cell immunity against S1 and S2 subunits were studied via IFN-γ enzyme-linked immunosorbent spot (ELISpot) and flow cytometric (FC) activation-induced marker (AIM) assays and the assessment of cytotoxic CD8+ T-cell function (in the subset of HLA-A2-positive patients). No single immunoassay was sufficient in identifying anti-spike convalescent immunity among all patients. There was no consistent correlation between adaptive humoral and cellular anti-spike responses. Our data indicate that the magnitude of anti-spike convalescent humoral and cellular immunity is highly heterogeneous and highlights the need for using multiple assays to comprehensively measure SARS-CoV-2 convalescent immunity. These observations might have implications for COVID-19 surveillance, and the determination of optimal vaccination strategies for emerging variants. Further studies are needed to determine the optimal assessment of adaptive humoral and cellular immunity following SARS-CoV-2 infection, especially in the context of emerging variants and unclear vaccination schedules.
Collapse
Affiliation(s)
- Balaji Pathakumari
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (B.P.); (P.K.M.); (M.S.); (V.P.V.K.); (P.A.-S.); (P.E.)
| | - Paige K. Marty
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (B.P.); (P.K.M.); (M.S.); (V.P.V.K.); (P.A.-S.); (P.E.)
| | - Maleeha Shah
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (B.P.); (P.K.M.); (M.S.); (V.P.V.K.); (P.A.-S.); (P.E.)
| | - Virginia P. Van Keulen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (B.P.); (P.K.M.); (M.S.); (V.P.V.K.); (P.A.-S.); (P.E.)
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA; (C.L.E.); (M.S.B.)
| | - Courtney L. Erskine
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA; (C.L.E.); (M.S.B.)
| | - Matthew S. Block
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA; (C.L.E.); (M.S.B.)
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Pedro Arias-Sanchez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (B.P.); (P.K.M.); (M.S.); (V.P.V.K.); (P.A.-S.); (P.E.)
| | - Patricio Escalante
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (B.P.); (P.K.M.); (M.S.); (V.P.V.K.); (P.A.-S.); (P.E.)
| | - Tobias Peikert
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (B.P.); (P.K.M.); (M.S.); (V.P.V.K.); (P.A.-S.); (P.E.)
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA; (C.L.E.); (M.S.B.)
| |
Collapse
|
29
|
Manirakiza A, Malaka C, Mossoro-Kpinde HD, Yambiyo BM, Mossoro-Kpinde CD, Fandema E, Niamathe Yakola C, Doyama-Woza R, Kangale-Wando IM, Kosh Komba JE, Nzapali Guiagassomon SMB, Namsenei-Dankpea LJVDLG, Coti-Reckoundji CSG, Bouhouda M, Gody JC, Grésenguet G, Vernet G, Vernet MA, Nakoune E. Seroprevalence of anti-SARS-CoV-2 antibodies before and after implementation of anti-COVID-19 vaccination among hospital staff in Bangui, Central African Republic. PLOS GLOBAL PUBLIC HEALTH 2023; 3:e0001497. [PMID: 37910467 PMCID: PMC10619860 DOI: 10.1371/journal.pgph.0001497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 10/09/2023] [Indexed: 11/03/2023]
Abstract
Healthcare workers (HCWs) are at high to very high risk for SARS-CoV-2 infection. The persistence of this pandemic worldwide has instigated the need for an investigation of the level of prevention through immunization and vaccination against SARS-CoV-2 among HCWs. The objective of our study was to evaluate any changes in anti-COVID-19 serological status before and after the vaccination campaign of health personnel in the Central African Republic. We carried out a repeated cross-sectional serological study on HCWs at the university hospital centers of Bangui. Blood samples were collected and tested for anti-SARS-CoV-2 IgM and IgG using the ELISA technique on blood samples. A total of 179 and 141 HCWs were included in the first and second surveys, respectively. Of these staff, 31.8% of HCWs were positive for anti-SARS-CoV-2 IgG in the first survey, whereas 95.7% were positive for anti-SARS-CoV-2 IgG in the second survey. However, the proportion of HCWs positive for SARS-CoV-2 IgM antibodies was low (9.7% in the first survey and 3.6% in the second survey). These findings showed a sharp increase in seroprevalence over a one-year period. This increase is primarily due to the synergistic effect of the infection and the implementation of vaccines against COVID-19. Further studies to assess the persistence of anti-SARS-CoV-2 antibodies are needed.
Collapse
Affiliation(s)
- Alexandre Manirakiza
- Institut Pasteur of Bangui, Pasteur International Network, Bangui, Central African Republic
- University of Bangui, Faculté des Sciences de la Santé, Bangui, Central African Republic
| | - Christian Malaka
- Institut Pasteur of Bangui, Pasteur International Network, Bangui, Central African Republic
| | | | - Brice Martial Yambiyo
- Institut Pasteur of Bangui, Pasteur International Network, Bangui, Central African Republic
- University of Bangui, Faculté des Sciences de la Santé, Bangui, Central African Republic
| | | | - Emmanuel Fandema
- University of Bangui, Faculté des Sciences de la Santé, Bangui, Central African Republic
| | | | - Rodrigue Doyama-Woza
- University of Bangui, Faculté des Sciences de la Santé, Bangui, Central African Republic
| | | | - Jess Elliot Kosh Komba
- Centre Hospitalier et Universitaire Pédiatrique de Bangui, Bangui, Central African Republic
| | | | | | | | - Modeste Bouhouda
- Institut Pasteur of Bangui, Pasteur International Network, Bangui, Central African Republic
| | - Jean-Chrisostome Gody
- University of Bangui, Faculté des Sciences de la Santé, Bangui, Central African Republic
- Centre Hospitalier et Universitaire Pédiatrique de Bangui, Bangui, Central African Republic
| | - Gérard Grésenguet
- University of Bangui, Faculté des Sciences de la Santé, Bangui, Central African Republic
| | - Guy Vernet
- Institut Pasteur of Bangui, Pasteur International Network, Bangui, Central African Republic
| | - Marie Astrid Vernet
- Institut Pasteur of Bangui, Pasteur International Network, Bangui, Central African Republic
| | - Emmanuel Nakoune
- Institut Pasteur of Bangui, Pasteur International Network, Bangui, Central African Republic
- University of Bangui, Faculté des Sciences de la Santé, Bangui, Central African Republic
| |
Collapse
|
30
|
Joshi D, Nyhoff LE, Zarnitsyna VI, Moreno A, Manning K, Linderman S, Burrell AR, Stephens K, Norwood C, Mantus G, Ahmed R, Anderson EJ, Staat MA, Suthar MS, Wrammert J. Infants and young children generate more durable antibody responses to SARS-CoV-2 infection than adults. iScience 2023; 26:107967. [PMID: 37822504 PMCID: PMC10562792 DOI: 10.1016/j.isci.2023.107967] [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: 07/28/2023] [Revised: 08/25/2023] [Accepted: 09/15/2023] [Indexed: 10/13/2023] Open
Abstract
As SARS-CoV-2 becomes endemic, it is critical to understand immunity following early-life infection. We evaluated humoral responses to SARS-CoV-2 in 23 infants/young children. Antibody responses to SARS-CoV-2 spike antigens peaked approximately 30 days after infection and were maintained up to 500 days with little apparent decay. While the magnitude of humoral responses was similar to an adult cohort recovered from mild/moderate COVID-19, both binding and neutralization titers to WT SARS-CoV-2 were more durable in infants/young children, with spike and RBD IgG antibody half-life nearly 4X as long as in adults. IgG subtype analysis revealed that while IgG1 formed the majority of the response in both groups, IgG3 was more common in adults and IgG2 in infants/young children. These findings raise important questions regarding differential regulation of humoral immunity in infants/young children and adults and could have broad implications for the timing of vaccination and booster strategies in this age group.
Collapse
Affiliation(s)
- Devyani Joshi
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
| | - Lindsay E. Nyhoff
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
| | | | - Alberto Moreno
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Emory National Primate Research Center, Atlanta, GA, USA
- Department of Medicine, Emory University, School of Medicine, Atlanta, GA, USA
| | - Kelly Manning
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Emory National Primate Research Center, Atlanta, GA, USA
| | - Susanne Linderman
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Allison R. Burrell
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Environmental and Public Health Sciences, Division of Epidemiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kathy Stephens
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
| | - Carson Norwood
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
| | - Grace Mantus
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
| | - Rafi Ahmed
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Evan J. Anderson
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
- Department of Medicine, Emory University, School of Medicine, Atlanta, GA, USA
| | - Mary A. Staat
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Mehul S. Suthar
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
- Emory National Primate Research Center, Atlanta, GA, USA
| | - Jens Wrammert
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
| |
Collapse
|
31
|
Munywoki PK, Bigogo G, Nasimiyu C, Ouma A, Aol G, Oduor CO, Rono S, Auko J, Agogo GO, Njoroge R, Oketch D, Odhiambo D, Odeyo VW, Kikwai G, Onyango C, Juma B, Hunsperger E, Lidechi S, Ochieng CA, Lo TQ, Munyua P, Herman-Roloff A. Heterogenous transmission and seroprevalence of SARS-CoV-2 in two demographically diverse populations with low vaccination uptake in Kenya, March and June 2021. Gates Open Res 2023; 7:101. [PMID: 37990692 PMCID: PMC10661969 DOI: 10.12688/gatesopenres.14684.2] [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] [Accepted: 09/29/2023] [Indexed: 11/23/2023] Open
Abstract
Background SARS-CoV-2 has extensively spread in cities and rural communities, and studies are needed to quantify exposure in the population. We report seroprevalence of SARS-CoV-2 in two well-characterized populations in Kenya at two time points. These data inform the design and delivery of public health mitigation measures. Methods Leveraging on existing population based infectious disease surveillance (PBIDS) in two demographically diverse settings, a rural site in western Kenya in Asembo, Siaya County, and an urban informal settlement in Kibera, Nairobi County, we set up a longitudinal cohort of randomly selected households with serial sampling of all consenting household members in March and June/July 2021. Both sites included 1,794 and 1,638 participants in the March and June/July 2021, respectively. Individual seroprevalence of SARS-CoV-2 antibodies was expressed as a percentage of the seropositive among the individuals tested, accounting for household clustering and weighted by the PBIDS age and sex distribution. Results Overall weighted individual seroprevalence increased from 56.2% (95%CI: 52.1, 60.2%) in March 2021 to 63.9% (95%CI: 59.5, 68.0%) in June 2021 in Kibera. For Asembo, the seroprevalence almost doubled from 26.0% (95%CI: 22.4, 30.0%) in March 2021 to 48.7% (95%CI: 44.3, 53.2%) in July 2021. Seroprevalence was highly heterogeneous by age and geography in these populations-higher seroprevalence was observed in the urban informal settlement (compared to the rural setting), and children aged <10 years had the lowest seroprevalence in both sites. Only 1.2% and 1.6% of the study participants reported receipt of at least one dose of the COVID-19 vaccine by the second round of serosurvey-none by the first round. Conclusions In these two populations, SARS-CoV-2 seroprevalence increased in the first 16 months of the COVID-19 pandemic in Kenya. It is important to prioritize additional mitigation measures, such as vaccine distribution, in crowded and low socioeconomic settings.
Collapse
Affiliation(s)
- Patrick K. Munywoki
- Division for Global Health Protection, Global Health Center, U.S. Centers for Disease Control and Prevention (CDC)-Kenya, Nairobi, Kenya
| | - Godfrey Bigogo
- Centre for Global Health Research, Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - Carolyne Nasimiyu
- Global Health Program, Washington State University – Global Health Kenya (WSU-GH Kenya), Nairobi, Kenya
- Paul G. Allen School of Global Health, Washington State University, Pullman, Washington, USA
| | - Alice Ouma
- Centre for Global Health Research, Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - George Aol
- Centre for Global Health Research, Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - Clifford O. Oduor
- Centre for Global Health Research, Kenya Medical Research Institute (KEMRI), Nairobi, Kenya
| | - Samuel Rono
- Centre for Global Health Research, Kenya Medical Research Institute (KEMRI), Nairobi, Kenya
| | - Joshua Auko
- Centre for Global Health Research, Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - George O. Agogo
- Division for Global Health Protection, Global Health Center, U.S. Centers for Disease Control and Prevention (CDC)-Kenya, Nairobi, Kenya
| | - Ruth Njoroge
- Global Health Program, Washington State University – Global Health Kenya (WSU-GH Kenya), Nairobi, Kenya
| | - Dismas Oketch
- Global Health Program, Washington State University – Global Health Kenya (WSU-GH Kenya), Nairobi, Kenya
| | - Dennis Odhiambo
- Centre for Global Health Research, Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - Victor W. Odeyo
- Centre for Global Health Research, Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - Gilbert Kikwai
- Centre for Global Health Research, Kenya Medical Research Institute (KEMRI), Nairobi, Kenya
| | - Clayton Onyango
- Division for Global Health Protection, Global Health Center, U.S. Centers for Disease Control and Prevention (CDC)-Kenya, Nairobi, Kenya
| | - Bonventure Juma
- Division for Global Health Protection, Global Health Center, U.S. Centers for Disease Control and Prevention (CDC)-Kenya, Nairobi, Kenya
| | - Elizabeth Hunsperger
- Division for Global Health Protection, Global Health Center, U.S. Centers for Disease Control and Prevention (CDC)-Kenya, Nairobi, Kenya
| | - Shirley Lidechi
- Centre for Global Health Research, Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | | | - Terrence Q. Lo
- Division for Global Health Protection, Global Health Center, U.S. Centers for Disease Control and Prevention (CDC)-Kenya, Nairobi, Kenya
| | - Peninah Munyua
- Division for Global Health Protection, Global Health Center, U.S. Centers for Disease Control and Prevention (CDC)-Kenya, Nairobi, Kenya
| | - Amy Herman-Roloff
- Division for Global Health Protection, Global Health Center, U.S. Centers for Disease Control and Prevention (CDC)-Kenya, Nairobi, Kenya
| |
Collapse
|
32
|
Abdullahi A, Frimpong J, Cheng MTK, Aliyu SH, Smith C, Abimiku A, Phillips RO, Owusu M, Gupta RK. Performance of SARS COV-2 IgG Anti-N as an Independent Marker of Exposure to SARS COV-2 in an Unvaccinated West African Population. Am J Trop Med Hyg 2023; 109:890-894. [PMID: 37580023 PMCID: PMC10551093 DOI: 10.4269/ajtmh.23-0179] [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: 03/22/2023] [Accepted: 05/22/2023] [Indexed: 08/16/2023] Open
Abstract
Determination of previous SARS-COV-2 infection is hampered by the absence of a standardized test. The marker used to assess previous exposure is IgG antibody to the nucleocapsid (IgG anti-N), although it is known to wane quickly from peripheral blood. The accuracies of seven antibody tests (virus neutralization test, IgG anti-N, IgG anti-spike [anti-S], IgG anti-receptor binding domain [anti-RBD], IgG anti-N + anti-RBD, IgG anti-N + anti-S, and IgG anti-S + anti-RBD), either singly or in combination, were evaluated on 502 cryopreserved serum samples collected before the COVID-19 vaccination rollout in Kumasi, Ghana. The accuracy of each index test was measured using a composite reference standard based on a combination of neutralization test and IgG anti-N antibody tests. According to the composite reference, 262 participants were previously exposed; the most sensitive test was the virus neutralization test, with 95.4% sensitivity (95% CI: 93.6-97.3), followed by 79.0% for IgG anti-N + anti-S (95% CI: 76.3-83.3). The most specific tests were virus neutralization and IgG anti-N, both with 100% specificity. Viral neutralization and IgG anti-N + anti-S were the overall most accurate tests, with specificity/sensitivity of 100/95.2% and 79.0/92.1%, respectively. Our findings indicate that IgG anti-N alone is an inadequate marker of prior exposure to SARS COV-2 in this population. Virus neutralization assay appears to be the most accurate assay in discerning prior infection. A combination of IgG anti-N and IgG anti-S is also accurate and suited for assessment of SARS COV-2 exposure in low-resource settings.
Collapse
Affiliation(s)
- Adam Abdullahi
- Cambridge Institute of Therapeutic Immunology & Infectious Disease, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Institute of Human Virology, Abuja, Nigeria
| | - James Frimpong
- Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Kumasi Centre for Collaborative Research in Tropical Medicine, Kumasi, Ghana
| | - Mark T. K. Cheng
- Cambridge Institute of Therapeutic Immunology & Infectious Disease, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Sani H. Aliyu
- Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | | | | | - Richard Odame Phillips
- Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Kumasi Centre for Collaborative Research in Tropical Medicine, Kumasi, Ghana
| | - Michael Owusu
- Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Kumasi Centre for Collaborative Research in Tropical Medicine, Kumasi, Ghana
| | - Ravindra K. Gupta
- Cambridge Institute of Therapeutic Immunology & Infectious Disease, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Africa Health Research Institute, Durban, South Africa
| |
Collapse
|
33
|
Miquel CH, Abbas F, Cenac C, Foret-Lucas C, Guo C, Ducatez M, Joly E, Hou B, Guéry JC. B cell-intrinsic TLR7 signaling is required for neutralizing antibody responses to SARS-CoV-2 and pathogen-like COVID-19 vaccines. Eur J Immunol 2023; 53:e2350437. [PMID: 37438976 DOI: 10.1002/eji.202350437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/16/2023] [Accepted: 06/13/2023] [Indexed: 07/14/2023]
Abstract
Toll-like receptor 7 (TLR7) triggers antiviral immune responses through its capacity to recognize single-stranded RNA. TLR7 loss-of-function mutants are associated with life-threatening pneumonia in severe COVID-19 patients. Whereas TLR7-driven innate induction of type I IFN appears central to control SARS-CoV2 virus spreading during the first days of infection, the impact of TLR7-deficiency on adaptive B-cell immunity is less clear. In the present study, we examined the role of TLR7 in the adaptive B cells response to various pathogen-like antigens (PLAs). We used inactivated SARS-CoV2 and a PLA-based COVID-19 vaccine candidate designed to mimic SARS-CoV2 with encapsulated bacterial ssRNA as TLR7 ligands and conjugated with the RBD of the SARS-CoV2 Spike protein. Upon repeated immunization with inactivated SARS-CoV2 or PLA COVID-19 vaccine, we show that Tlr7-deficiency abolished the germinal center (GC)-dependent production of RBD-specific class-switched IgG2b and IgG2c, and neutralizing antibodies to SARS-CoV2. We also provide evidence for a non-redundant role for B-cell-intrinsic TLR7 in the promotion of RBD-specific IgG2b/IgG2c and memory B cells. Together, these data demonstrate that the GC reaction and class-switch recombination to the Myd88-dependent IgG2b/IgG2c in response to SARS-CoV2 or PLAs is strictly dependent on cell-intrinsic activation of TLR7 in B cells.
Collapse
Affiliation(s)
- Charles-Henry Miquel
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (INFINITY), Université de Toulouse, INSERM, CNRS, UPS, Toulouse, France
- Arthritis R&D, Neuilly-Sur-Seine, France
| | - Flora Abbas
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (INFINITY), Université de Toulouse, INSERM, CNRS, UPS, Toulouse, France
| | - Claire Cenac
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (INFINITY), Université de Toulouse, INSERM, CNRS, UPS, Toulouse, France
| | - Charlotte Foret-Lucas
- Interactions Hôtes Agents Pathogènes (IHAP), UMR1225, Université de Toulouse, INRAe, ENVT, Toulouse, France
| | - Chang Guo
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Mariette Ducatez
- Interactions Hôtes Agents Pathogènes (IHAP), UMR1225, Université de Toulouse, INRAe, ENVT, Toulouse, France
| | - Etienne Joly
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Baidong Hou
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Jean-Charles Guéry
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (INFINITY), Université de Toulouse, INSERM, CNRS, UPS, Toulouse, France
| |
Collapse
|
34
|
Leomanni L, Collatuzzo G, Sansone E, Sala E, De Palma G, Porru S, Spiteri G, Monaco MGL, Basso D, Pavanello S, Scapellato ML, Larese Filon F, Cegolon L, Mauro M, Lodi V, Lazzarotto T, Noreña I, Reinkemeyer C, Giang LTT, Fabiánová E, Strhársky J, Dell’Omo M, Murgia N, Carrasco-Ribelles LA, Violán C, Mates D, Rascu A, Vimercati L, De Maria L, Asafo SS, Ditano G, Abedini M, Boffetta P. Determinants of Anti-S Immune Response at 12 Months after SARS-CoV-2 Vaccination in a Multicentric European Cohort of Healthcare Workers-ORCHESTRA Project. Vaccines (Basel) 2023; 11:1527. [PMID: 37896931 PMCID: PMC10610704 DOI: 10.3390/vaccines11101527] [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: 08/02/2023] [Revised: 09/18/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND The effectiveness of the immunity provided by SARS-CoV-2 vaccines is an important public health issue. We analyzed the determinants of 12-month serology in a multicenter European cohort of vaccinated healthcare workers (HCW). METHODS We analyzed the sociodemographic characteristics and levels of anti-SARS-CoV-2 spike antibodies (IgG) in a cohort of 16,101 vaccinated HCW from eleven centers in Germany, Italy, Romania, Slovakia and Spain. Considering the skewness of the distribution, the serological levels were transformed using log or cubic standardization and normalized by dividing them by center-specific standard errors. We fitted center-specific multivariate regression models to estimate the cohort-specific relative risks (RR) of an increase of one standard deviation of log or cubic antibody level and the corresponding 95% confidence interval (CI) for different factors and combined them in random-effects meta-analyses. RESULTS We included 16,101 HCW in the analysis. A high antibody level was positively associated with age (RR = 1.04, 95% CI = 1.00-1.08 per 10-year increase), previous infection (RR = 1.78, 95% CI 1.29-2.45) and use of Spikevax [Moderna] with combinations compared to Comirnaty [BioNTech/Pfizer] (RR = 1.07, 95% CI 0.97-1.19) and was negatively associated with the time since last vaccine (RR = 0.94, 95% CI 0.91-0.98 per 30-day increase). CONCLUSIONS These results provide insight about vaccine-induced immunity to SARS-CoV-2, an analysis of its determinants and quantification of the antibody decay trend with time since vaccination.
Collapse
Affiliation(s)
- Ludovica Leomanni
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (L.L.)
| | - Giulia Collatuzzo
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (L.L.)
| | - Emanuele Sansone
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, 25121 Brescia, Italy
| | - Emma Sala
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, 25121 Brescia, Italy
| | - Giuseppe De Palma
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, 25121 Brescia, Italy
| | - Stefano Porru
- Section of Occupational Medicine, Department of Diagnostics and Public Health, University of Verona, 37129 Verona, Italy
- Clinical Unit of Occupational Medicine, University Hospital of Verona, 37100 Verona, Italy
| | - Gianluca Spiteri
- Clinical Unit of Occupational Medicine, University Hospital of Verona, 37100 Verona, Italy
| | | | - Daniela Basso
- Department of Medicine-DIMED, University of Padova, 35128 Padova, Italy
- Laboratory Medicine Unit, University Hospital of Padova, 35128 Padova, Italy
| | - Sofia Pavanello
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
- Occupational Medicine Unit, University Hospital of Padova, 35128 Padova, Italy
| | - Maria Luisa Scapellato
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
- Occupational Medicine Unit, University Hospital of Padova, 35128 Padova, Italy
| | - Francesca Larese Filon
- Occupational Medicine Unit, Department of Medicine, Surgery and Health Sciences, University of Trieste, 34100 Trieste, Italy
| | - Luca Cegolon
- Occupational Medicine Unit, Department of Medicine, Surgery and Health Sciences, University of Trieste, 34100 Trieste, Italy
| | - Marcella Mauro
- Occupational Medicine Unit, Department of Medicine, Surgery and Health Sciences, University of Trieste, 34100 Trieste, Italy
| | - Vittorio Lodi
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (L.L.)
- SSD Health Surveillance, IRCCS University Hospital, 40139 Bologna, Italy
| | - Tiziana Lazzarotto
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (L.L.)
- Microbiology Unit, IRCCS University Hospital, 40139 Bologna, Italy
| | - Ivan Noreña
- Division of Infectious Diseases and Tropical Medicine, LMU University Hospital, 81377 Munich, Germany
| | - Christina Reinkemeyer
- Division of Infectious Diseases and Tropical Medicine, LMU University Hospital, 81377 Munich, Germany
| | - Le Thi Thu Giang
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, LMU University Hospital, 81377 Munich, Germany
| | - Eleonóra Fabiánová
- Occupational Health Department, Regional Authority of Public Health, 497556 Banská Bystrica, Slovakia
| | - Jozef Strhársky
- Medical Microbiology Department, Regional Authority of Public Health, 497556 Banská Bystrica, Slovakia
| | - Marco Dell’Omo
- Unit of Occupational Medicine, Department on Medicine and Surgery, University of Perugia, 06125 Perugia, Italy
| | - Nicola Murgia
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Lucía A. Carrasco-Ribelles
- Unitat de Suport a la Recerca Metropolitana Nord, Institut Universitari d’Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), 08303 Mataró, Spain
- Direcció d’Atenció Primària Metropolitana Nord Institut Català de Salut, 08007 Barcelona, Spain
- Grup de Recerca en Impacte de les Malalties Cròniques i les seves Trajectòries (GRIMTra), Institut Universitari d’Investigació en Atenció Primària Jordi Gol (IDIAPJGol), 08303 Barcelona, Spain
- Network for Research on Chronicity, Primary Care and Health Promotion (RICAPPS), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Concepción Violán
- Unitat de Suport a la Recerca Metropolitana Nord, Institut Universitari d’Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), 08303 Mataró, Spain
- Direcció d’Atenció Primària Metropolitana Nord Institut Català de Salut, 08007 Barcelona, Spain
- Grup de Recerca en Impacte de les Malalties Cròniques i les seves Trajectòries (GRIMTra), Institut Universitari d’Investigació en Atenció Primària Jordi Gol (IDIAPJGol), 08303 Barcelona, Spain
- Network for Research on Chronicity, Primary Care and Health Promotion (RICAPPS), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
- Universitat Autónoma de Barcelona, 08193 Bellaterra, Spain
| | - Dana Mates
- National Institute of Public Health, 050463 Bucharest, Romania
| | - Agripina Rascu
- Department of Internal Medicine-Occupational Medicine, Medicine and Pharmacy “Carol Davila”, 050474 Bucharest, Romania
| | - Luigi Vimercati
- Interdisciplinary Department of Medicine, University of Bari, 70121 Bari, Italy
| | - Luigi De Maria
- Interdisciplinary Department of Medicine, University of Bari, 70121 Bari, Italy
| | - Shuffield S. Asafo
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (L.L.)
| | - Giorgia Ditano
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (L.L.)
| | - Mahsa Abedini
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (L.L.)
| | - Paolo Boffetta
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (L.L.)
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA
- Department of Family, Population and Preventive Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
35
|
Corrado MM, Jia X, Geno Rasmussen C, Pyle L, Yu L, Liu E, Stahl M, Rewers MJ. Previous SARS-CoV-2 Infection Is Not Associated With Increased Celiac Disease Autoimmunity in Children and Adolescents. Am J Gastroenterol 2023; 118:1698-1700. [PMID: 37159249 DOI: 10.14309/ajg.0000000000002317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/27/2023] [Indexed: 05/10/2023]
Abstract
INTRODUCTION Recent reports suggest severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infections may increase the risk of celiac disease autoimmunity. This study aims to evaluate potential associations between coronavirus disease 2019 infection and tissue transglutaminase autoantibodies (TGA) immunoglobulin A. METHODS From 2020 to 2021, cross-sectional screening for SARS-CoV-2 antibodies and TGA was offered to 4,717 children in Colorado through the Autoimmunity Screening for Kids study. Multivariable logistic regression assessed association between previous SARS-CoV-2 infection and TGA positivity. RESULTS Previous SARS-CoV-2 infection was not associated with TGA positivity (odds ratio 1.02, 95% confidence interval 0.63-1.59; P = 0.95). DISCUSSION In this large-scale analysis, previous SARS-CoV-2 infection was not associated with celiac disease autoimmunity in Colorado children.
Collapse
Affiliation(s)
- Michelle M Corrado
- Digestive Health Institute, Children's Hospital Colorado, University of Colorado, Aurora, Colorado, USA
| | - Xiaofan Jia
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, Colorado, USA
| | | | - Laura Pyle
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, Colorado, USA
| | - Liping Yu
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, Colorado, USA
| | - Edwin Liu
- Digestive Health Institute, Children's Hospital Colorado, University of Colorado, Aurora, Colorado, USA
| | - Marisa Stahl
- Digestive Health Institute, Children's Hospital Colorado, University of Colorado, Aurora, Colorado, USA
| | - Marian J Rewers
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
36
|
Rouvinski A, Friedman A, Kirillov S, Attal JH, Kumari S, Fahoum J, Wiener R, Magen S, Plotkin Y, Chemtob D, Bercovier H. Antibody response in elderly vaccinated four times with an mRNA anti-COVID-19 vaccine. Sci Rep 2023; 13:14165. [PMID: 37644113 PMCID: PMC10465611 DOI: 10.1038/s41598-023-41399-5] [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: 04/09/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023] Open
Abstract
The humoral response after the fourth dose of a mRNA vaccine against COVID-19 has not been adequately described in elderly recipients, particularly those not exposed previously to SARS-CoV-2. Serum anti-RBD IgG levels (Abbott SARS-CoV-2 IgG II Quant assay) and neutralizing capacities (spike SARS-CoV-2 pseudovirus Wuhan and Omicron BA.1 variant) were measured after the third and fourth doses of a COVID-19 mRNA vaccine among 46 elderly residents (median age 85 years [IQR 81; 89]) of an assisted living facility. Among participants never infected by SARS-CoV-2, the mean serum IgG levels against RBD (2025 BAU/ml), 99 days after the fourth vaccine, was as high as 76 days after the third vaccine (1987 BAU/ml), and significantly higher (p = 0.030) when the latter were corrected for elapsed time. Neutralizing antibody levels against the historical Wuhan strain were significantly higher (Mean 1046 vs 1573; p = 0.002) and broader (against Omicron) (Mean 170 vs 375; p = 0.018), following the fourth vaccine. The six individuals with an Omicron breakthrough infection mounted strong immune responses for anti-RBD and neutralizing antibodies against the Omicron variant indicating that the fourth vaccine dose did not prevent a specific adaptation of the immune response. These findings point out the value of continued vaccine boosting in the elderly population.
Collapse
Affiliation(s)
- Alexander Rouvinski
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Ahuva Friedman
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Saveliy Kirillov
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of General Biology and Genomics, L.N. Gumilyov Eurasian National University, Astana, Kazakhstan
| | - Jordan Hannink Attal
- Braun School of Public Health and Community Medicine, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Tuberculosis and AIDS, State of Israel Ministry of Health, Jerusalem, Israel
| | - Sujata Kumari
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jamal Fahoum
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Reuven Wiener
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sophie Magen
- Department of Clinical Biochemistry, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Yevgeni Plotkin
- Department of Anesthesiology, Critical Care and Pain Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Daniel Chemtob
- Braun School of Public Health and Community Medicine, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Tuberculosis and AIDS, State of Israel Ministry of Health, Jerusalem, Israel
| | - Herve Bercovier
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
37
|
Marty PK, Pathakumari B, Shah M, Keulen VP, Erskine CL, Block MS, Arias-Sanchez P, Escalante P, Peikert T. Convalescent Adaptive Immunity is Highly Heterogenous after SARS-CoV-2 Infection. RESEARCH SQUARE 2023:rs.3.rs-3222112. [PMID: 37674707 PMCID: PMC10479471 DOI: 10.21203/rs.3.rs-3222112/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Optimal detection strategies for effective convalescent immunity after SARS-CoV-2 infection and vaccination remain unclear. The objective of this study was to characterize convalescent immunity targeting the SARS-CoV-2 spike protein using a multiparametric approach. At the beginning of the pandemic, between April 23, 2020, to May 11, 2020, we recruited 30 COVID-19 unvaccinated convalescent donors and 7 unexposed asymptomatic donors. Peripheral blood mononuclear cells (PBMCs) were obtained from leukapheresis cones. The humoral immune response was assessed by measuring serum anti-SARS-CoV-2 spike S1 subunit IgG semiquantitative ELISA and T cell immunity against S1 and S2 subunits were studied by IFN-γ Enzyme-Linked Immune absorbent Spot (ELISpot), flow cytometric (FC) activation-induced marker (AIM) assays and the assessment of cytotoxic CD8+ T-cell function (in the subset of HLA-A2 positive patients). No single immunoassay was sufficient in identifying anti-spike convalescent immunity among all patients. There was no consistent correlation between adaptive humoral and cellular anti-spike responses. Our data indicate that the magnitude of anti-spike convalescent humoral and cellular immunity is highly heterogeneous and highlights the need for using multiple assays to comprehensively measure SARS-CoV-2 convalescent immunity. These observations might have implications for COVID-19 surveillance, and optimal vaccination strategies for emerging variants. Further studies are needed to determine the optimal assessment of adaptive humoral and cellular immunity following SARSCoV-2 infection, especially in the context of emerging variants and unclear vaccination schedules.
Collapse
|
38
|
de Gier B, Huiberts AJ, Hoeve CE, den Hartog G, van Werkhoven H, van Binnendijk R, Hahné SJM, de Melker HE, van den Hof S, Knol MJ. Effects of COVID-19 vaccination and previous infection on Omicron SARS-CoV-2 infection and relation with serology. Nat Commun 2023; 14:4793. [PMID: 37558656 PMCID: PMC10412579 DOI: 10.1038/s41467-023-40195-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/17/2023] [Indexed: 08/11/2023] Open
Abstract
An increasing proportion of the population has acquired immunity through COVID-19 vaccination and previous SARS-CoV-2 infection, i.e., hybrid immunity, possibly affecting the risk of new infection. We aim to estimate the protective effect of previous infections and vaccinations on SARS-CoV-2 Omicron infection, using data from 43,257 adult participants in a prospective community-based cohort study in the Netherlands, collected between 10 January 2022 and 1 September 2022. Our results show that, for participants with 2, 3 or 4 prior immunizing events (vaccination or previous infection), hybrid immunity is more protective against infection with SARS-CoV-2 Omicron than vaccine-induced immunity, up to at least 30 weeks after the last immunizing event. Differences in risk of infection are partly explained by differences in anti-Spike RBD (S) antibody concentration, which is associated with risk of infection in a dose-response manner. Among participants with hybrid immunity, with one previous pre-Omicron infection, we do not observe a relevant difference in risk of Omicron infection by sequence of vaccination(s) and infection. Additional immunizing events increase the protection against infection, but not above the level of the first weeks after the previous event.
Collapse
Affiliation(s)
- Brechje de Gier
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Anne J Huiberts
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Christina E Hoeve
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Gerco den Hartog
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Henri van Werkhoven
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Rob van Binnendijk
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Susan J M Hahné
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Hester E de Melker
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Susan van den Hof
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Mirjam J Knol
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands.
| |
Collapse
|
39
|
Gromowski GD, Cincotta CM, Mayer S, King J, Swafford I, McCracken MK, Coleman D, Enoch J, Storme C, Darden J, Peel S, Epperson D, McKee K, Currier JR, Okulicz J, Paquin-Proulx D, Cowden J, Peachman K. Humoral immune responses associated with control of SARS-CoV-2 breakthrough infections in a vaccinated US military population. EBioMedicine 2023; 94:104683. [PMID: 37413891 PMCID: PMC10345251 DOI: 10.1016/j.ebiom.2023.104683] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND COVID-19 vaccines have been critical for protection against severe disease following infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) but gaps remain in our understanding of the immune responses that contribute to controlling subclinical and mild infections. METHODS Vaccinated, active-duty US military service members were enrolled in a non-interventional, minimal-risk, observational study starting in May, 2021. Clinical data, serum, and saliva samples were collected from study participants and were used to characterise the humoral immune responses to vaccination and to assess its impact on clinical and subclinical infections, as well as virologic outcomes of breakthrough infections (BTI) including viral load and infection duration. FINDINGS The majority of VIRAMP participants had received the Pfizer COVID-19 vaccine and by January, 2022, N = 149 had a BTI. The median BTI duration (PCR+ days) was 4 days and the interquartile range was 1-8 days. Participants that were nucleocapsid seropositive prior to their BTI had significantly higher levels of binding and functional antibodies to the spike protein, shorter median duration of infections, and lower median peak viral loads compared to seronegative participants. Furthermore, levels of neutralising antibody, ACE2 blocking activity, and spike-specific IgA measured prior to BTI also correlated with the duration of infection. INTERPRETATION We extended previous findings and demonstrate that a subset of vaccine-induced humoral immune responses, along with nucleocapsid serostatus are associated with control of SARS-CoV-2 breakthrough infections in the upper airways. FUNDING This work was funded by the DoD Joint Program Executive Office for Chemical, Biological, Radiological and Nuclear Defense (JPEO-CBRND) in collaboration with the Defense Health Agency (DHA) COVID-19 funding initiative for the VIRAMP study.
Collapse
Affiliation(s)
- Gregory D Gromowski
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA.
| | - Camila Macedo Cincotta
- Diagnostics and Countermeasures Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Sandra Mayer
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA; Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Jocelyn King
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA; Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Isabella Swafford
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA; U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Michael K McCracken
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Dante Coleman
- Diagnostics and Countermeasures Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Jennifer Enoch
- Diagnostics and Countermeasures Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Casey Storme
- Diagnostics and Countermeasures Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Janice Darden
- Diagnostics and Countermeasures Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Sheila Peel
- Diagnostics and Countermeasures Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Diane Epperson
- Booz Allen Hamilton, McLean, VA, USA; Enabling Biotechnologies, Joint Program Executive Office for Chemical, Biological, Radiological and Nuclear Defense, Frederick, MD, USA
| | | | - Jeffrey R Currier
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Jason Okulicz
- Department of Infectious Disease, Brooke Army Medical Center, San Antonio, TX, USA
| | - Dominic Paquin-Proulx
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA; U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Jessica Cowden
- Enabling Biotechnologies, Joint Program Executive Office for Chemical, Biological, Radiological and Nuclear Defense, Frederick, MD, USA; Department of Retrovirology, U.S. Army Medical Directorate-Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand.
| | - Kristina Peachman
- Diagnostics and Countermeasures Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| |
Collapse
|
40
|
Seekircher L, Bánki Z, Kimpel J, Rössler A, Schäfer H, Falkensammer B, Bante D, Forer L, Schönherr S, Harthaller T, Sacher M, Ower C, Tschiderer L, Ulmer H, Krammer F, von Laer D, Borena W, Willeit P. Immune response after two doses of the BNT162b2 COVID-19 vaccine and risk of SARS-CoV-2 breakthrough infection in Tyrol, Austria: an open-label, observational phase 4 trial. THE LANCET. MICROBE 2023; 4:e612-e621. [PMID: 37354911 PMCID: PMC10284585 DOI: 10.1016/s2666-5247(23)00107-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 02/03/2023] [Accepted: 03/06/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND Correlates of protection could help to assess the extent to which a person is protected from SARS-CoV-2 infection after vaccination (so-called breakthrough infection). We aimed to clarify associations of antibody and T-cell responses after vaccination against COVID-19 with risk of a SARS-CoV-2 breakthrough infection and whether measurement of these responses enhances risk prediction. METHODS We did an open-label, phase 4 trial in two community centres in the Schwaz district of the Federal State of Tyrol, Austria, before the emergence of the omicron (B.1.1.529) variant of SARS-CoV-2. We included individuals (aged ≥16 years) a mean of 35 days (range 27-43) after they had received a second dose of the BNT162b2 (Pfizer-BioNTech) COVID-19 vaccine. We quantified associations between immunological parameters and breakthrough infection and assessed whether information on these parameters improves risk discrimination. The study is registered with the European Union Drug Regulating Authorities Clinical Trials Database, 2021-002030-16. FINDINGS 2760 individuals (1682 [60·9%] female, 1078 [39·1%] male, mean age 47·4 years [SD 14·5]) were enrolled into this study between May 15 and May 21, 2021, 712 (25·8%) of whom had a previous SARS-CoV-2 infection. Over a median follow-up of 5·9 months, 68 (2·5%) participants had a breakthrough infection. In models adjusted for age, sex, and previous infection, hazard ratios for breakthrough infection for having twice the immunological parameter level at baseline were 0·72 (95% CI 0·60-0·86) for anti-spike IgG, 0·80 (0·70-0·92) for neutralising antibodies in a surrogate virus neutralisation assay, 0·84 (0·58-1·21) for T-cell response after stimulation with a CD4 peptide pool, and 0·77 (0·54-1·08) for T-cell response after stimulation with a combined CD4 and CD8 peptide pool. For neutralising antibodies measured in a nested case-control sample using a pseudotyped virus neutralisation assay, the corresponding odds ratio was 0·78 (0·62-1·00). Among participants with previous infection, the corresponding hazard ratio was 0·73 (0·61-0·88) for anti-nucleocapsid Ig. Addition of anti-spike IgG information to a model containing information on age and sex improved the C-index by 0·085 (0·027-0·143). INTERPRETATION In contrast to T-cell response, higher levels of binding and neutralising antibodies were associated with a reduced risk of breakthrough SARS-CoV-2 infection. The assessment of anti-spike IgG enhances the prediction of incident breakthrough SARS-CoV-2 infection and could therefore be a suitable correlate of protection in practice. Our phase 4 trial measured both humoral and cellular immunity and had a 6-month follow-up period; however, the longer-term protection against emerging variants of SARS-CoV-2 remains unclear. FUNDING None.
Collapse
Affiliation(s)
- Lisa Seekircher
- Institute of Health Economics, Medical University of Innsbruck, Innsbruck, Austria
| | - Zoltán Bánki
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Janine Kimpel
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Annika Rössler
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Helena Schäfer
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - David Bante
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas Forer
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sebastian Schönherr
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Teresa Harthaller
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Magdalena Sacher
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Cornelia Ower
- Department of Surgery, University Hospital of Trauma Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Lena Tschiderer
- Institute of Health Economics, Medical University of Innsbruck, Innsbruck, Austria
| | - Hanno Ulmer
- Institute of Medical Statistics and Informatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Krammer
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; 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
| | - Dorothee von Laer
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Wegene Borena
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria.
| | - Peter Willeit
- Institute of Health Economics, Medical University of Innsbruck, Innsbruck, Austria; Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Vienna, Austria; Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
| |
Collapse
|
41
|
Invernizzi A, Renzetti S, van Thriel C, Rechtman E, Patrono A, Ambrosi C, Mascaro L, Cagna G, Gasparotti R, Reichenberg A, Tang CY, Lucchini RG, Wright RO, Placidi D, Horton MK. Covid-19 related cognitive, structural and functional brain changes among Italian adolescents and young adults: a multimodal longitudinal case-control study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.07.19.23292909. [PMID: 37503251 PMCID: PMC10371098 DOI: 10.1101/2023.07.19.23292909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Coronavirus disease 2019 (COVID-19) has been associated with brain functional, structural, and cognitive changes that persist months after infection. Most studies of the neurologic outcomes related to COVID-19 focus on severe infection and aging populations. Here, we investigated the neural activities underlying COVID-19 related outcomes in a case-control study of mildly infected youth enrolled in a longitudinal study in Lombardy, Italy, a global hotspot of COVID-19. All participants (13 cases, 27 controls, mean age 24 years) completed resting state functional (fMRI), structural MRI, cognitive assessments (CANTAB spatial working memory) at baseline (pre-COVID) and follow-up (post-COVID). Using graph theory eigenvector centrality (EC) and data-driven statistical methods, we examined differences in ECdelta (i.e., the difference in EC values pre- and post-COVID-19) and volumetricdelta (i.e., the difference in cortical volume of cortical and subcortical areas pre- and post-COVID) between COVID-19 cases and controls. We found that ECdeltasignificantly between COVID-19 and healthy participants in five brain regions; right intracalcarine cortex, right lingual gyrus, left hippocampus, left amygdala, left frontal orbital cortex. The left hippocampus showed a significant decrease in volumetricdelta between groups (p=0.041). The reduced ECdelta in the right amygdala associated with COVID-19 status mediated the association between COVID-19 and disrupted spatial working memory. Our results show persistent structural, functional and cognitive brain changes in key brain areas associated with olfaction and cognition. These results may guide treatment efforts to assess the longevity, reversibility and impact of the observed brain and cognitive changes following COVID-19.
Collapse
Affiliation(s)
- Azzurra Invernizzi
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stefano Renzetti
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Christoph van Thriel
- Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
| | - Elza Rechtman
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alessandra Patrono
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Claudia Ambrosi
- Department of Neuroscience, Neuroradiology Unit, ASST Cremona
| | | | - Giuseppa Cagna
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Roberto Gasparotti
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Abraham Reichenberg
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Cheuk Y Tang
- Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Roberto G Lucchini
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, United States
| | - Robert O Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Donatella Placidi
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Megan K Horton
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
42
|
Amellal H, Assaid N, Charoute H, Akarid K, Maaroufi A, Ezzikouri S, Sarih M. Kinetics of specific anti-SARS-CoV-2 IgM, IgA, and IgG responses during the first 12 months after SARS-CoV-2 infection: A prospective longitudinal study. PLoS One 2023; 18:e0288557. [PMID: 37437051 DOI: 10.1371/journal.pone.0288557] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 06/29/2023] [Indexed: 07/14/2023] Open
Abstract
Coronavirus 2019 (COVID-19) is a global health threat. The kinetics of antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) need to be assessed, as the long-term duration of these immunoglobulins remains largely controversial. The aim of this study was to assess the longitudinal dynamics of anti-SARS-CoV-2 antibodies against the nucleocapsid (N) protein and the receptor-binding domain (RBD) of the spike protein up to one year in a cohort of 190 COVID-19 patients. Between March and September 2021, we enrolled patients from two regional hospitals in Casablanca, Morocco. Blood samples were collected and analyzed for antibody levels. We used the commercial Euroimmun ELISA for the determination of anti-N IgM, the Abbott Architect™ SARS-CoV-2 IgG test for the detection of anti-RBD IgG, and an in-house kit for the assay of anti-N IgG and anti-N IgA. IgM and IgA antibodies were assessed 2-5, 9-12, 17-20 and 32-37 days after symptom onset. IgG antibodies were also assessed 60, 90, 120 and 360 days after symptom onset. One-third of patients developed IgM (32%), while two-thirds developed IgA (61%). One month of symptom onset, most patients developed IgG, with 97% and 93% positivity for anti-RBD IgG and anti-N IgG, respectively. The anti-RBD IgG positivity rate remained high up to one year of follow-up. However, the anti-N IgG positivity rate decreased over time, with only 41% of patients testing positive after one year's follow-up. IgG levels were significantly higher in older people (over 50 years) than in other study participants. We also found that patients who had received two doses of ChAdOx1 nCoV-19 vaccine prior to infection had a lower IgM response than unvaccinated patients. This difference was statistically significant two weeks after the onset of symptoms. We present the first study in Africa to measure the kinetics of antibody response (IgA, IgM and IgG) to SARS-CoV-2 over one year. Most participants remained seropositive for anti-RBD IgG after one year but showed a significant decline in antibody titers.
Collapse
Affiliation(s)
- Houda Amellal
- Department of Parasitology and Vector-Borne Diseases, Institut Pasteur du Maroc, Casablanca, Morocco
- Aïn Chock Faculty of Sciences, Health and Environment Laboratory, Biochemistry, Biotechnology and Immunophysiopathology Research Team, Hassan II University of Casablanca, Casablanca, Morocco
| | - Najlaa Assaid
- Department of Parasitology and Vector-Borne Diseases, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Hicham Charoute
- Institut Pasteur du Maroc, Research Unit of Epidemiology, Biostatistics and Bioinformatics, Casablanca, Morocco
| | - Khadija Akarid
- Aïn Chock Faculty of Sciences, Health and Environment Laboratory, Biochemistry, Biotechnology and Immunophysiopathology Research Team, Hassan II University of Casablanca, Casablanca, Morocco
| | - Abderrahmane Maaroufi
- Department of Parasitology and Vector-Borne Diseases, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Sayeh Ezzikouri
- Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Virology Unit, Casablanca, Morocco
| | - M'hammed Sarih
- Department of Parasitology and Vector-Borne Diseases, Institut Pasteur du Maroc, Casablanca, Morocco
| |
Collapse
|
43
|
Xu J, Zheng J, Tan Y, Cai J, Xiang Y, Ling H, Li Z, Bai Q. Longitudinal Observation of Immune Response for 23 Months in COVID-19 Convalescent Patients After Infection and Vaccination. Viral Immunol 2023; 36:389-400. [PMID: 37276049 DOI: 10.1089/vim.2022.0111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023] Open
Abstract
To better understand dynamic changes of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) immune response, a prospective, single-center, cohort study was conducted on longitudinal immune response in 34 COVID-19 convalescent patients over 23 months in Chongqing. Two blood samples from convalescent patients were collected, first sample collected during 10-13 months (M10-13) after infection (pre-SARS-CoV-2 vaccination) and second sample collected during 20-23 months (M20-23) after infection (post-SARS-CoV-2 vaccination). The SARS-CoV-2-specific humoral and cellular immunity were traced by testing total antibody (Ab), anti-nucleocapsid (NP) immunoglobulin M (IgM), anti-NP immunoglobulin G (IgG), and anti-spike (S) IgG Abs, lymphocyte subset count, and Th1 cytokines. Healthy donors (30) were also included in the study as the uninspected healthy controls. Our data showed significant change in mean titer of SARS-CoV-2-specific Ab response from M10-13 to M20-23 included, namely, SARS-CoV-2-specific total Ab as 219 AU/mL increasing to 750.9 AU/mL; anti-NP IgM as 3.5 AU/mL decreasing significantly (p < 0.001) to 0.6 AU/mL; anti-NP IgG as 7.9 AU/mL increasing to 87.1 AU/mL; and anti-S IgG as 499.0 RU/mL increasing to 1,802.3 RU/mL. Our observations suggested that one vaccine dose might have been sufficient for COVID-19 convalescent patients. Larger sample sizes are needed to compare better immune effect of protein subunit vaccine. Besides, compared to healthy donors, patients had decreased CD3+ and CD8+ T lymphocyte counts during two periods. Patients had most cytokines recovered normally within 2 years, but IL-6 level was significantly elevated; however, IL-6 was negatively correlated with IgM and positively correlated with IgG. Changes in cytokines might have been caused by SARS-CoV-2 infection or vaccination. Patients with comorbidities were associated with decreased CD3+ and CD8+ T lymphocytes and lower Ab titers following SARS-CoV-2 vaccination. Vaccination enormously increased humoral immunity beneficial in COVID-19 convalescent patients. Elderly COVID-19 convalescent patients with comorbidities needed more attention.
Collapse
Affiliation(s)
- Jingru Xu
- College of Public Health, Chongqing Medical University, Chongqing, China
- Microbiological Laboratory, Chongqing Center for Disease Control and Prevention, Chongqing, China
| | - Juan Zheng
- Department of Neurology, Chongqing Red Cross Hospital (People's Hospital of Jiangbei District), Chongqing, China
| | - Yan Tan
- Microbiological Laboratory, Chongqing Center for Disease Control and Prevention, Chongqing, China
| | - Jiaojiao Cai
- Microbiological Laboratory, Chongqing Center for Disease Control and Prevention, Chongqing, China
| | - Yao Xiang
- Microbiological Laboratory, Chongqing Center for Disease Control and Prevention, Chongqing, China
| | - Hua Ling
- Microbiological Laboratory, Chongqing Center for Disease Control and Prevention, Chongqing, China
| | - Zhifeng Li
- Microbiological Laboratory, Chongqing Center for Disease Control and Prevention, Chongqing, China
| | - Qunhua Bai
- College of Public Health, Chongqing Medical University, Chongqing, China
| |
Collapse
|
44
|
Simons E, Nikolay B, Ouedraogo P, Pasquier E, Tiemeni C, Adjaho I, Badjo C, Chamman K, Diomandé M, Dosso M, Doumbia M, Izia YA, Kakompe H, Katsomya AM, Kij V, Akissi VK, Mambula C, Mbala-Kingebeni P, Muzinga J, Ngoy B, Penali L, Pini A, Porten K, Salou H, Sevede D, Luquero F, Gignoux E. Seroprevalence of SARS-CoV-2 antibodies and retrospective mortality in two African settings: Lubumbashi, Democratic Republic of the Congo and Abidjan, Côte d'Ivoire. PLOS GLOBAL PUBLIC HEALTH 2023; 3:e0001457. [PMID: 37289736 DOI: 10.1371/journal.pgph.0001457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 05/10/2023] [Indexed: 06/10/2023]
Abstract
Although seroprevalence studies have demonstrated the wide circulation of SARS-COV-2 in African countries, the impact on population health in these settings is still poorly understood. Using representative samples of the general population, we evaluated retrospective mortality and seroprevalence of anti-SARS-CoV-2 antibodies in Lubumbashi and Abidjan. The studies included retrospective mortality surveys and nested anti-SARS-CoV-2 antibody prevalence surveys. In Lubumbashi the study took place during April-May 2021 and in Abidjan the survey was implemented in two phases: July-August 2021 and October-November 2021. Crude mortality rates were stratified between pre-pandemic and pandemic periods and further investigated by age group and COVID waves. Anti-SARS-CoV-2 seroprevalence was quantified by rapid diagnostic testing (RDT) and laboratory-based testing (ELISA in Lubumbashi and ECLIA in Abidjan). In Lubumbashi, the crude mortality rate (CMR) increased from 0.08 deaths per 10 000 persons per day (pre-pandemic) to 0.20 deaths per 10 000 persons per day (pandemic period). Increases were particularly pronounced among <5 years old. In Abidjan, no overall increase was observed during the pandemic period (pre-pandemic: 0.05 deaths per 10 000 persons per day; pandemic: 0.07 deaths per 10 000 persons per day). However, an increase was observed during the third wave (0.11 deaths per 10 000 persons per day). The estimated seroprevalence in Lubumbashi was 15.7% (RDT) and 43.2% (laboratory-based). In Abidjan, the estimated seroprevalence was 17.4% (RDT) and 72.9% (laboratory-based) during the first phase of the survey and 38.8% (RDT) and 82.2% (laboratory-based) during the second phase of the survey. Although circulation of SARS-CoV-2 seems to have been extensive in both settings, the public health impact varied. The increases, particularly among the youngest age group, suggest indirect impacts of COVID and the pandemic on population health. The seroprevalence results confirmed substantial underdetection of cases through the national surveillance systems.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Hugues Kakompe
- Ministry of Health, Kinshasa, Democratic Republic of the Congo
| | | | - Vicky Kij
- Ministry of Health, Kinshasa, Democratic Republic of the Congo
| | | | | | | | - Jacques Muzinga
- Laboratoire National de Lubumbashi, Lubumbashi, Democratic Republic of the Congo
| | - Basile Ngoy
- Ministry of Health, Kinshasa, Democratic Republic of the Congo
| | - Lou Penali
- Institut Pasteur Cote d'Ivoire, Abidjan, Cote d'Ivoire
| | | | | | | | - Daouda Sevede
- Institut Pasteur Cote d'Ivoire, Abidjan, Cote d'Ivoire
| | | | | |
Collapse
|
45
|
DeJonge PM, Lambrou AS, Segaloff HE, Bateman A, Sterkel A, Griggs C, Baggott J, Kelly P, Thornburg N, Epperson M, Desamu-Thorpe R, Abedi G, Hsu CH, Nakayama JY, Ruffin J, Turner-Harper D, Matanock A, Almendares O, Whaley M, Chakrabarti A, DeGruy K, Daly M, Westergaard R, Tate JE, Kirking HL. Assessment of Anti-SARS-CoV-2 antibody levels among university students vaccinated with different COVID-19 primary and booster doses - fall 2021, Wisconsin. BMC Infect Dis 2023; 23:374. [PMID: 37277736 DOI: 10.1186/s12879-023-08332-7] [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: 01/23/2023] [Accepted: 05/16/2023] [Indexed: 06/07/2023] Open
Abstract
BACKGROUND University students commonly received COVID-19 vaccinations before returning to U.S. campuses in the Fall of 2021. Given likely immunologic variation among students based on differences in type of primary series and/or booster dose vaccine received, we conducted serologic investigations in September and December 2021 on a large university campus in Wisconsin to assess anti-SARS-CoV-2 antibody levels. METHODS We collected blood samples, demographic information, and COVID-19 illness and vaccination history from a convenience sample of students. Sera were analyzed for both anti-spike (anti-S) and anti-nucleocapsid (anti-N) antibody levels using World Health Organization standardized binding antibody units per milliliter (BAU/mL). Levels were compared across categorical primary COVID-19 vaccine series received and binary COVID-19 mRNA booster status. The association between anti-S levels and time since most recent vaccination dose was estimated by mixed-effects linear regression. RESULTS In total, 356 students participated, of whom 219 (61.5%) had received a primary vaccine series of Pfizer-BioNTech or Moderna mRNA vaccines and 85 (23.9%) had received vaccines from Sinovac or Sinopharm. Median anti-S levels were significantly higher for mRNA primary vaccine series recipients (2.90 and 2.86 log [BAU/mL], respectively), compared with those who received Sinopharm or Sinovac vaccines (1.63 and 1.95 log [BAU/mL], respectively). Sinopharm and Sinovac vaccine recipients were associated with a significantly faster anti-S decline over time, compared with mRNA vaccine recipients (P <.001). By December, 48/172 (27.9%) participants reported receiving an mRNA COVID-19 vaccine booster, which reduced the anti-S antibody discrepancies between primary series vaccine types. CONCLUSIONS Our work supports the benefit of heterologous boosting against COVID-19. COVID-19 mRNA vaccine booster doses were associated with increases in anti-SARS-CoV-2 antibody levels; following an mRNA booster dose, students with both mRNA and non-mRNA primary series receipt were associated with comparable levels of anti-S IgG.
Collapse
Affiliation(s)
- Peter M DeJonge
- Epidemic Intelligence Service, CDC, Atlanta, Georgia, 30329, USA.
- Wisconsin Department of Health Services, Division of Public Health, Madison, Wisconsin, 53703, USA.
| | | | - Hannah E Segaloff
- Epidemic Intelligence Service, CDC, Atlanta, Georgia, 30329, USA
- Wisconsin Department of Health Services, Division of Public Health, Madison, Wisconsin, 53703, USA
| | - Allen Bateman
- Wisconsin State Laboratory of Hygiene, Madison, Wisconsin, 53703, USA
| | - Alana Sterkel
- Wisconsin State Laboratory of Hygiene, Madison, Wisconsin, 53703, USA
| | - Carol Griggs
- University Health Services, University of Wisconsin - Madison, Madison, Wisconsin, 53703, USA
| | - Jake Baggott
- University Health Services, University of Wisconsin - Madison, Madison, Wisconsin, 53703, USA
| | - Patrick Kelly
- University Health Services, University of Wisconsin - Madison, Madison, Wisconsin, 53703, USA
| | | | | | | | - Glen Abedi
- CDC COVID-19 Response Team, Atlanta, Georgia, 30329, USA
| | | | - Jasmine Y Nakayama
- Epidemic Intelligence Service, CDC, Atlanta, Georgia, 30329, USA
- CDC COVID-19 Response Team, Atlanta, Georgia, 30329, USA
| | - Jasmine Ruffin
- CDC COVID-19 Response Team, Atlanta, Georgia, 30329, USA
| | | | - Almea Matanock
- CDC COVID-19 Response Team, Atlanta, Georgia, 30329, USA
| | | | - Melissa Whaley
- CDC COVID-19 Response Team, Atlanta, Georgia, 30329, USA
| | | | - Kyle DeGruy
- CDC COVID-19 Response Team, Atlanta, Georgia, 30329, USA
| | - Michele Daly
- CDC COVID-19 Response Team, Atlanta, Georgia, 30329, USA
| | - Ryan Westergaard
- Wisconsin Department of Health Services, Division of Public Health, Madison, Wisconsin, 53703, USA
| | | | | |
Collapse
|
46
|
Bannick MS, Gao F, Brown ER, Janes HE. Retrospective, Observational Studies for Estimating Vaccine Effects on the Secondary Attack Rate of SARS-CoV-2. Am J Epidemiol 2023; 192:1016-1028. [PMID: 36883907 PMCID: PMC10505422 DOI: 10.1093/aje/kwad046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 11/21/2022] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) vaccines are highly efficacious at preventing symptomatic infection, severe disease, and death. Most of the evidence that COVID-19 vaccines also reduce transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is based on retrospective, observational studies. Specifically, an increasing number of studies are evaluating vaccine effectiveness against the secondary attack rate of SARS-CoV-2 using data available in existing health-care databases or contact-tracing databases. Since these types of databases were designed for clinical diagnosis or management of COVID-19, they are limited in their ability to provide accurate information on infection, infection timing, and transmission events. We highlight challenges with using existing databases to identify transmission units and confirm potential SARS-CoV-2 transmission events. We discuss the impact of common diagnostic testing strategies, including event-prompted and infrequent testing, and illustrate their potential biases in estimating vaccine effectiveness against the secondary attack rate of SARS-CoV-2. We articulate the need for prospective observational studies of vaccine effectiveness against the SARS-CoV-2 secondary attack rate, and we provide design and reporting considerations for studies using retrospective databases.
Collapse
Affiliation(s)
- Marlena S Bannick
- Correspondence to Marlena Bannick, Department of Biostatistics, Hans Rosling Center for Population Health, Box 357232, University of Washington, Seattle, WA 98195 (e-mail: )
| | | | | | | |
Collapse
|
47
|
Patalon T, Saciuk Y, Perez G, Peretz A, Ben-Tov A, Gazit S. Dynamics of Naturally Acquired Immunity Against Severe Acute Respiratory Syndrome Coronavirus 2 in Children and Adolescents. J Pediatr 2023; 257:113371. [PMID: 36870558 PMCID: PMC9981270 DOI: 10.1016/j.jpeds.2023.02.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 02/03/2023] [Accepted: 02/12/2023] [Indexed: 03/06/2023]
Abstract
OBJECTIVE To evaluate the duration of protection against reinfection conferred by a previous severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in children and adolescents. STUDY DESIGN We applied 2 complementary approaches: a matched test-negative, case-control design and a retrospective cohort design. A total of 458 959 unvaccinated individuals aged 5-18 years were included. The analyses focused on the period July 1, 2021, to December 13, 2021, a period of Delta variant dominance in Israel. We evaluated 3 SARS-CoV-2-related outcomes: documented polymerase chain reaction-confirmed infection or reinfection, symptomatic infection or reinfection, and SARS-CoV-2-related hospitalization or death. RESULTS Overall, children and adolescents who were previously infected acquired durable protection against reinfection with SARS-CoV-2 for at least 18 months. Importantly, no SARS-CoV-2-related deaths were recorded in either the SARS-CoV-2-naïve group or the previously infected group. The effectiveness of naturally acquired immunity against a recurrent infection reached 89.2% (95% CI, 84.7%-92.4%) at 3-6 months after the first infection and declined slightly to 82.5% (95% CI, 79.1%-85.3%) by 9-12 months after infection, with a slight nonsignificant waning trend seen up to 18 months after infection. Additionally, children aged 5-11 years exhibited no significant waning of naturally acquired protection throughout the outcome period, whereas waning protection in those aged 12-18 years was more prominent but still mild. CONCLUSIONS Children and adolescents who were previously infected with SARS-CoV-2 remain protected to a high degree for 18 months. Further research is needed to examine naturally acquired immunity against Omicron and newer emerging variants.
Collapse
Affiliation(s)
- Tal Patalon
- Kahn Sagol Maccabi Research & Innovation Center, Maccabi Healthcare Services, Tel Aviv, Israel; Maccabitech Institute for Research and Innovation, Maccabi Healthcare Services, Tel Aviv, Israel.
| | - Yaki Saciuk
- Kahn Sagol Maccabi Research & Innovation Center, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Galit Perez
- Maccabitech Institute for Research and Innovation, Maccabi Healthcare Services, Tel Aviv, Israel; Department of Health Policy and Management, School of Public Health, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Asaf Peretz
- Maccabitech Institute for Research and Innovation, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Amir Ben-Tov
- Kahn Sagol Maccabi Research & Innovation Center, Maccabi Healthcare Services, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sivan Gazit
- Kahn Sagol Maccabi Research & Innovation Center, Maccabi Healthcare Services, Tel Aviv, Israel; Maccabitech Institute for Research and Innovation, Maccabi Healthcare Services, Tel Aviv, Israel
| |
Collapse
|
48
|
Joshi D, Nyhoff LE, Zarnitsyna VI, Moreno A, Manning K, Linderman S, Burrell AR, Stephens K, Norwood C, Mantus G, Ahmed R, Anderson EJ, Staat MA, Suthar MS, Wrammert J. Infants and young children generate more durable antibody responses to SARS-CoV-2 infection than adults. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.10.23288360. [PMID: 37090559 PMCID: PMC10120804 DOI: 10.1101/2023.04.10.23288360] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Since the emergence of SARS-CoV-2, research has shown that adult patients mount broad and durable immune responses to infection. However, response to infection remains poorly studied in infants/young children. In this study, we evaluated humoral responses to SARS-CoV-2 in 23 infants/young children before and after infection. We found that antibody responses to SARS-CoV-2 spike antigens peaked approximately 30 days after infection and were maintained up to 500 days with little apparent decay. While the magnitude of humoral responses was similar to an adult cohort recovered from mild/moderate COVID-19, both binding and neutralization titers to WT SARS-CoV-2 were more durable in infants/young children, with Spike and RBD IgG antibody half-life nearly 4X as long as in adults. The functional breadth of adult and infant/young children SARS-CoV-2 responses were comparable, with similar reactivity against panel of recent and previously circulating viral variants. Notably, IgG subtype analysis revealed that while IgG1 formed the majority of both adults' and infants/young children's response, IgG3 was more common in adults and IgG2 in infants/young children. These findings raise important questions regarding differential regulation of humoral immunity in infants/young children and adults and could have broad implications for the timing of vaccination and booster strategies in this age group.
Collapse
|
49
|
Kosiorek P, Stróż S, Hryniewicz A, Kazberuk DE, Milewski R, Bartoszewicz K, Borkowska MJ, Stasiak‐Barmuta A. A new set-up of vanishing antibodies: A biennial follow-up of five different clients' humoral responses against SARS-CoV-2 after systemic vaccination in an oncology hospital in Poland. Health Sci Rep 2023; 6:e1172. [PMID: 37008810 PMCID: PMC10064024 DOI: 10.1002/hsr2.1172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/13/2023] [Indexed: 04/03/2023] Open
Affiliation(s)
- Piotr Kosiorek
- Department of EmergencyMaria Sklodowska‐Curie Bialystok Oncology CentreBiałystokPoland
- Department of Clinical ImmunologyMedical University of BiałystokBiałystokPoland
| | - Samuel Stróż
- Department of Clinical ImmunologyMedical University of BiałystokBiałystokPoland
| | - Anna Hryniewicz
- Department of RehabilitationMedical University of BiałystokBiałystokPoland
| | - Dorota E. Kazberuk
- Department of RadiotherapyMaria Sklodowska‐Curie Bialystok Oncology CentreBiałystokPoland
| | - Robert Milewski
- Department of Biostatistics and Medical InformaticsMedical University of BiałystokBiałystokPoland
| | | | - Magdalena J. Borkowska
- Department of RadiotherapyMaria Sklodowska‐Curie Bialystok Oncology CentreBiałystokPoland
| | | |
Collapse
|
50
|
Saavedra C, Vallejo A, Longo F, Serrano JJ, Fernández M, Gion M, López-Miranda E, Martínez-Jáñez N, Guerra E, Chamorro J, Rosero D, Velasco H, Martín A, Carrato A, Casado JL, Cortés A. Discordant Humoral and T-Cell Response to mRNA SARS-CoV-2 Vaccine and the Risk of Breakthrough Infections in Women with Breast Cancer, Receiving Cyclin-Dependent Kinase 4 and 6 Inhibitors. Cancers (Basel) 2023; 15:cancers15072000. [PMID: 37046661 PMCID: PMC10093435 DOI: 10.3390/cancers15072000] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/09/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
Few data are available about the immune response to mRNA SARS-CoV-2 vaccines in patients with breast cancer receiving cyclin-dependent kinase 4/6 inhibitors (CDK4/6i). We conducted a prospective, single-center study of patients with breast cancer treated with CDK4/6i who received mRNA-1273 vaccination, as well as a comparative group of healthcare workers. The primary endpoint was to compare the rate and magnitude of humoral and T-cell response after full vaccination. A better neutralizing antibody and anti-S IgG level was observed after vaccination in the subgroup of women receiving CDK4/6i, but a trend toward a reduced CD4 and CD8 T-cell response in the CDK4/6i group was not statistically significant. There were no differences in the rate of COVID-19 after vaccination (19% vs. 12%), but breakthrough infections were observed in those with lower levels of anti-S IgG and neutralizing antibodies after the first dose. A lower rate of CD4 T-cell response was also found in those individuals with breakthrough infections, although a non-significant and similar level of CD8 T-cell response was also observed, regardless of breakthrough infections. The rate of adverse events was higher in patients treated with CDK4/6i, without serious adverse events. In conclusion, there was a robust humoral response, but a blunted T-cell response to mRNA vaccine in women receiving CDK4/6i, suggesting a reduced trend of the adaptative immune response.
Collapse
Affiliation(s)
- Cristina Saavedra
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - Alejandro Vallejo
- Laboratory of Immunovirology, Department of Infectious Diseases, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - Federico Longo
- CIBERONC, Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, Alcalá University, 28034 Madrid, Spain
| | - Juan José Serrano
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - María Fernández
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - María Gion
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - Elena López-Miranda
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - Noelia Martínez-Jáñez
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - Eva Guerra
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - Jesús Chamorro
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - Diana Rosero
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - Héctor Velasco
- CiberInfect, Infectious Disease Department, Ramón y Cajal University Hospital, 28034 Madrid, Spain
| | - Adrián Martín
- CiberInfect, Infectious Disease Department, Ramón y Cajal University Hospital, 28034 Madrid, Spain
| | - Alfredo Carrato
- CIBERONC, Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, Alcalá University, 28034 Madrid, Spain
| | - José Luis Casado
- CiberInfect, Infectious Disease Department, Ramón y Cajal University Hospital, 28034 Madrid, Spain
- Correspondence: (J.L.C.); (A.C.)
| | - Alfonso Cortés
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
- Correspondence: (J.L.C.); (A.C.)
| |
Collapse
|