651
|
Meyers LM, Gutiérrez AH, Boyle CM, Terry F, McGonnigal BG, Salazar A, Princiotta MF, Martin WD, De Groot AS, Moise L. Highly conserved, non-human-like, and cross-reactive SARS-CoV-2 T cell epitopes for COVID-19 vaccine design and validation. NPJ Vaccines 2021; 6:71. [PMID: 33986292 PMCID: PMC8119491 DOI: 10.1038/s41541-021-00331-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/01/2021] [Indexed: 12/24/2022] Open
Abstract
Natural and vaccine-induced SARS-CoV-2 immunity in humans has been described but correlates of protection are not yet defined. T cells support the SARS-CoV-2 antibody response, clear virus-infected cells, and may be required to block transmission. In this study, we identified peptide epitopes associated with SARS-CoV-2 T-cell immunity. Using immunoinformatic methods, T-cell epitopes from spike, membrane, and envelope were selected for maximal HLA-binding potential, coverage of HLA diversity, coverage of circulating virus, and minimal potential cross-reactivity with self. Direct restimulation of PBMCs collected from SARS-CoV-2 convalescents confirmed 66% of predicted epitopes, whereas only 9% were confirmed in naive individuals. However, following a brief period of epitope-specific T-cell expansion, both cohorts demonstrated robust T-cell responses to 97% of epitopes. HLA-DR3 transgenic mouse immunization with peptides co-formulated with poly-ICLC generated a potent Th1-skewed, epitope-specific memory response, alleviating safety concerns of enhanced respiratory disease associated with Th2 induction. Taken together, these epitopes may be used to improve our understanding of natural and vaccine-induced immunity, and to facilitate the development of T-cell-targeted vaccines that harness pre-existing SARS-CoV-2 immunity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Anne S De Groot
- EpiVax, Inc., Providence, RI, USA.
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA.
| | - Leonard Moise
- EpiVax, Inc., Providence, RI, USA.
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA.
- Institute for Immunology and Immunoinformatics, University of Rhode Island, Providence, RI, USA.
| |
Collapse
|
652
|
Abstract
PURPOSE OF REVIEW Despite its crucial role in protection against viral infections, mucosal immunity has been largely understudied in the context of coronavirus disease 2019 (COVID-19). This review outlines the current evidence about the role of mucosal immune responses in the clearance of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, as well as potential mucosal mechanisms of protection against (re-)infection. RECENT FINDINGS The angiotensin-converting enzyme 2 cellular entry receptor for SARS-CoV-2 is most highly expressed in the upper respiratory tract and most SARS-CoV-2 shedding occurs from the upper respiratory tract. Viral shedding peaks early during infection around the onset of symptoms, before dropping rapidly in most individuals within 7 days of symptom onset, suggesting mucosal inhibition of viral infection. Serum and mucosal immunoglobulin G and immunoglobulin M responses were found to be strongly correlated in infected patients, whereas correlations were much weaker for immunoglobulin A (IgA). Mucosal IgA responses have been detected in infected cases in the absence of serum antibody responses, with mucosal antibody levels correlating strongly with virus neutralization. Bulk and single-cell RNA sequencing analysis of nasopharyngeal swabs and bronchoalveolar lavage samples of COVID-19 patients revealed the induction of mucosal chemokine and cytokine genes, complement pathways, Janus Kinase/Signal Transducer and Activator of Transcription signaling and cytotoxic T cells. SUMMARY Although most clinical studies focus on antibodies and cellular immunity in peripheral blood, mucosal immune responses in the respiratory tract play a key role in the early restriction of viral replication and the clearance of SARS-CoV-2. Identification of mucosal biomarkers associated with viral clearance will allow monitoring of infection-induced immunity. Further studies are needed to understand how the systemic immunological endpoints measured in vaccination studies translate to mucosal protection against SARS-CoV-2 infection.
Collapse
|
653
|
Jahrsdörfer B, Groß R, Seidel A, Wettstein L, Ludwig C, Schwarz T, Körper S, Rojewski M, Lotfi R, Weinstock C, Seifried E, Corman VM, Drosten C, Münch J, Schrezenmeier H. Characterization of the SARS-CoV-2 Neutralization Potential of COVID-19-Convalescent Donors. THE JOURNAL OF IMMUNOLOGY 2021; 206:2614-2622. [PMID: 33980583 DOI: 10.4049/jimmunol.2100036] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022]
Abstract
The current SARS-CoV-2 pandemic has triggered the development of various SARS-CoV-2 neutralization tests. A wild-type virus (using African green monkey VeroE6 cells), a pseudovirus (using human Caco-2 cells), and a surrogate neutralization test platform were applied to characterize the SARS-CoV-2 neutralization potential of a cohort of 111 convalescent plasma donors over a period of seven months after diagnosis. This allowed an in-depth validation and assay performance analysis of these platforms. More importantly, we found that SARS-CoV-2 neutralization titers were stable or even increased within the observation period, which contradicts earlier studies reporting a rapid waning of Ab titers after three to four months. Moreover, we observed a positive correlation of neutralization titers with increasing age, number of symptoms reported, and the presence of the Rhesus Ag RhD. Validation of the platforms revealed that highest assay performances were obtained with the wild-type virus and the surrogate neutralization platforms. However, our data also suggested that selection of cutoff titers had a strong impact on the evaluation of neutralization potency. When taking strong neutralization potency, as demonstrated by the wild-type virus platform as the gold standard, up to 55% of plasma products had low neutralization titers. However, a significant portion of these products were overrated in their potency when using the surrogate assay with the recommended cutoff titer. In summary, our study demonstrates that SARS-CoV-2 neutralization titers are stable for at least seven months after diagnosis and offers a testing strategy for rapid selection of high-titer convalescent plasma products in a biosafety level 1 environment.
Collapse
Affiliation(s)
- Bernd Jahrsdörfer
- Department of Transfusion Medicine, Ulm University, Ulm, Germany; .,Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, Ulm, Germany
| | - Rüdiger Groß
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Alina Seidel
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Lukas Wettstein
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Carolin Ludwig
- Department of Transfusion Medicine, Ulm University, Ulm, Germany.,Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, Ulm, Germany
| | - Tatjana Schwarz
- Institute of Virology, Charité Universitätsmedizin Berlin, Berlin, Germany; and
| | - Sixten Körper
- Department of Transfusion Medicine, Ulm University, Ulm, Germany.,Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, Ulm, Germany
| | - Markus Rojewski
- Department of Transfusion Medicine, Ulm University, Ulm, Germany.,Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, Ulm, Germany
| | - Ramin Lotfi
- Department of Transfusion Medicine, Ulm University, Ulm, Germany.,Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, Ulm, Germany
| | - Christoph Weinstock
- Department of Transfusion Medicine, Ulm University, Ulm, Germany.,Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, Ulm, Germany
| | - Erhard Seifried
- Institute of Transfusion Medicine and Immunohematology, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen, Frankfurt, Germany
| | - Victor Max Corman
- Institute of Virology, Charité Universitätsmedizin Berlin, Berlin, Germany; and
| | - Christian Drosten
- Institute of Virology, Charité Universitätsmedizin Berlin, Berlin, Germany; and
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Hubert Schrezenmeier
- Department of Transfusion Medicine, Ulm University, Ulm, Germany.,Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, Ulm, Germany
| |
Collapse
|
654
|
Mishra PK, Bruiners N, Ukey R, Datta P, Onyuka A, Handler D, Hussain S, Honnen W, Singh S, Guerrini V, Yin Y, Dewald H, Choudhary A, Horton DB, Barrett ES, Roy J, Weiss SH, Fitzgerald-Bocarsly P, Blaser MJ, Carson JL, Panettieri RA, Lardizabal A, Chang TLY, Pinter A, Gennaro ML. Vaccination boosts protective responses and counters SARS-CoV-2-induced pathogenic memory B cells. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021. [PMID: 33880486 PMCID: PMC8057254 DOI: 10.1101/2021.04.11.21255153] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Much is to be learned about the interface between immune responses to SARS-CoV-2 infection and vaccination. We monitored immune responses specific to SARS-CoV-2 Spike Receptor-Binding-Domain (RBD) in convalescent individuals for eight months after infection diagnosis and following vaccination. Over time, neutralizing antibody responses, which are predominantly RBD specific, generally decreased, while RBD-specific memory B cells persisted. RBD-specific antibody and B cell responses to vaccination were more vigorous than those elicited by infection in the same subjects or by vaccination in infection-naïve comparators. Notably, the frequencies of double negative B memory cells, which are dysfunctional and potentially pathogenic, increased in the convalescent subjects over time. Unexpectedly, this effect was reversed by vaccination. Our work identifies a novel aspect of immune dysfunction in mild/moderate COVID-19, supports the practice of offering SARS-CoV-2 vaccination regardless of infection history, and provides a potential mechanistic explanation for the vaccination-induced reduction of “Long-COVID” symptoms.
Collapse
Affiliation(s)
- Pankaj Kumar Mishra
- Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Natalie Bruiners
- Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Rahul Ukey
- Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Pratik Datta
- Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Alberta Onyuka
- Global Tuberculosis Institute, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Deborah Handler
- Global Tuberculosis Institute, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Sabiha Hussain
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901
| | - William Honnen
- Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Sukhwinder Singh
- NJMS Flow Cytometry and Immunology Core Laboratory, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Valentina Guerrini
- Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Yue Yin
- Center for Advanced Biotechnology and Medicine, School of Public Health, Rutgers University, Piscataway, NJ 08854
| | - Hannah Dewald
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Alok Choudhary
- Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Daniel B Horton
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901
| | - Emily S Barrett
- Environmental and Occupational Health Sciences Institute, School of Public Health, Rutgers University, Piscataway, NJ 08854.,Department of Biostatistics and Epidemiology, School of Public Health, Rutgers University, Piscataway, NJ 08854
| | - Jason Roy
- Department of Biostatistics and Epidemiology, School of Public Health, Rutgers University, Piscataway, NJ 08854
| | - Stanley H Weiss
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103
| | | | - Martin J Blaser
- Center for Advanced Biotechnology and Medicine, School of Public Health, Rutgers University, Piscataway, NJ 08854
| | - Jeffrey L Carson
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901
| | | | - Alfred Lardizabal
- Global Tuberculosis Institute, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Theresa Li-Yun Chang
- Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Abraham Pinter
- Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Maria Laura Gennaro
- Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ 07103
| |
Collapse
|
655
|
Goupil R, Benlarbi M, Beaubien-Souligny W, Nadeau-Fredette AC, Chatterjee D, Goyette G, Gunaratnam L, Lamarche C, Tom A, Finzi A, Suri RS. Short-term antibody response after 1 dose of BNT162b2 vaccine in patients receiving hemodialysis. CMAJ 2021; 193:E793-E800. [PMID: 33980499 PMCID: PMC8177936 DOI: 10.1503/cmaj.210673] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND: Patients receiving in-centre hemodialysis are at high risk of exposure to SARS-CoV-2 and death if infected. One dose of the BNT162b2 SARS-CoV-2 vaccine is efficacious in the general population, but responses in patients receiving hemodialysis are uncertain. METHODS: We obtained serial plasma from patients receiving hemodialysis and health care worker controls before and after vaccination with 1 dose of the BNT162b2 mRNA vaccine, as well as convalescent plasma from patients receiving hemodialysis who survived COVID-19. We measured anti–receptor binding domain (RBD) immunoglobulin G (IgG) levels and stratified groups by evidence of previous SARS-CoV-2 infection. RESULTS: Our study included 154 patients receiving hemodialysis (135 without and 19 with previous SARS-CoV-2 infection), 40 controls (20 without and 20 with previous SARS-CoV-2 infection) and convalescent plasma from 16 patients. Among those without previous SARS-CoV-2 infection, anti-RBD IgG was undetectable at 4 weeks in 75 of 131 (57%, 95% confidence interval [CI] 47% to 65%) patients receiving hemodialysis, compared with 1 of 20 (5%, 95% CI 1% to 23%) controls (p < 0.001). No patient with nondetectable levels at 4 weeks developed anti-RBD IgG by 8 weeks. Results were similar in non-immunosuppressed and younger individuals. Three patients receiving hemodialysis developed severe COVID-19 after vaccination. Among those with previous SARS-CoV-2 infection, median anti-RBD IgG levels at 8 weeks in patients receiving hemodialysis were similar to controls at 3 weeks (p = 0.3) and to convalescent plasma (p = 0.8). INTERPRETATION: A single dose of BNT162b2 vaccine failed to elicit a humoral immune response in most patients receiving hemodialysis without previous SARS-CoV-2 infection, even after prolonged observation. In those with previous SARS-CoV-2 infection, the antibody response was delayed. We advise that patients receiving hemodialysis be prioritized for a second BNT162b2 dose at the recommended 3-week interval.
Collapse
Affiliation(s)
- Rémi Goupil
- Centre de recherche de l'Hôpital du Sacré-Cœur de Montréal (Goupil); Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM) (Benlarbi, Chatterjee, Goyette, Finzi, Suri); Centre de recherche de l'Hôpital Maisonneuve-Rosemont (Nadeau-Fredette, Lamarche), Montréal, Que.; Department of Microbiology and Immunology and Division of Nephrology, Department of Medicine (Gunaratnam), Western University, London, Ont.; Research Institute of the McGill University Health Centre (Tom, Suri); Département de microbiologie, infectiologie et immunologie (Finzi), Université de Montréal; Department of Microbiology and Immunology (Finzi), McGill University; Division of Nephrology (Suri), Department of Medicine, McGill University, Montréal, Que
| | - Mehdi Benlarbi
- Centre de recherche de l'Hôpital du Sacré-Cœur de Montréal (Goupil); Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM) (Benlarbi, Chatterjee, Goyette, Finzi, Suri); Centre de recherche de l'Hôpital Maisonneuve-Rosemont (Nadeau-Fredette, Lamarche), Montréal, Que.; Department of Microbiology and Immunology and Division of Nephrology, Department of Medicine (Gunaratnam), Western University, London, Ont.; Research Institute of the McGill University Health Centre (Tom, Suri); Département de microbiologie, infectiologie et immunologie (Finzi), Université de Montréal; Department of Microbiology and Immunology (Finzi), McGill University; Division of Nephrology (Suri), Department of Medicine, McGill University, Montréal, Que
| | - William Beaubien-Souligny
- Centre de recherche de l'Hôpital du Sacré-Cœur de Montréal (Goupil); Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM) (Benlarbi, Chatterjee, Goyette, Finzi, Suri); Centre de recherche de l'Hôpital Maisonneuve-Rosemont (Nadeau-Fredette, Lamarche), Montréal, Que.; Department of Microbiology and Immunology and Division of Nephrology, Department of Medicine (Gunaratnam), Western University, London, Ont.; Research Institute of the McGill University Health Centre (Tom, Suri); Département de microbiologie, infectiologie et immunologie (Finzi), Université de Montréal; Department of Microbiology and Immunology (Finzi), McGill University; Division of Nephrology (Suri), Department of Medicine, McGill University, Montréal, Que
| | - Annie-Claire Nadeau-Fredette
- Centre de recherche de l'Hôpital du Sacré-Cœur de Montréal (Goupil); Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM) (Benlarbi, Chatterjee, Goyette, Finzi, Suri); Centre de recherche de l'Hôpital Maisonneuve-Rosemont (Nadeau-Fredette, Lamarche), Montréal, Que.; Department of Microbiology and Immunology and Division of Nephrology, Department of Medicine (Gunaratnam), Western University, London, Ont.; Research Institute of the McGill University Health Centre (Tom, Suri); Département de microbiologie, infectiologie et immunologie (Finzi), Université de Montréal; Department of Microbiology and Immunology (Finzi), McGill University; Division of Nephrology (Suri), Department of Medicine, McGill University, Montréal, Que
| | - Debashree Chatterjee
- Centre de recherche de l'Hôpital du Sacré-Cœur de Montréal (Goupil); Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM) (Benlarbi, Chatterjee, Goyette, Finzi, Suri); Centre de recherche de l'Hôpital Maisonneuve-Rosemont (Nadeau-Fredette, Lamarche), Montréal, Que.; Department of Microbiology and Immunology and Division of Nephrology, Department of Medicine (Gunaratnam), Western University, London, Ont.; Research Institute of the McGill University Health Centre (Tom, Suri); Département de microbiologie, infectiologie et immunologie (Finzi), Université de Montréal; Department of Microbiology and Immunology (Finzi), McGill University; Division of Nephrology (Suri), Department of Medicine, McGill University, Montréal, Que
| | - Guillaume Goyette
- Centre de recherche de l'Hôpital du Sacré-Cœur de Montréal (Goupil); Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM) (Benlarbi, Chatterjee, Goyette, Finzi, Suri); Centre de recherche de l'Hôpital Maisonneuve-Rosemont (Nadeau-Fredette, Lamarche), Montréal, Que.; Department of Microbiology and Immunology and Division of Nephrology, Department of Medicine (Gunaratnam), Western University, London, Ont.; Research Institute of the McGill University Health Centre (Tom, Suri); Département de microbiologie, infectiologie et immunologie (Finzi), Université de Montréal; Department of Microbiology and Immunology (Finzi), McGill University; Division of Nephrology (Suri), Department of Medicine, McGill University, Montréal, Que
| | - Lakshman Gunaratnam
- Centre de recherche de l'Hôpital du Sacré-Cœur de Montréal (Goupil); Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM) (Benlarbi, Chatterjee, Goyette, Finzi, Suri); Centre de recherche de l'Hôpital Maisonneuve-Rosemont (Nadeau-Fredette, Lamarche), Montréal, Que.; Department of Microbiology and Immunology and Division of Nephrology, Department of Medicine (Gunaratnam), Western University, London, Ont.; Research Institute of the McGill University Health Centre (Tom, Suri); Département de microbiologie, infectiologie et immunologie (Finzi), Université de Montréal; Department of Microbiology and Immunology (Finzi), McGill University; Division of Nephrology (Suri), Department of Medicine, McGill University, Montréal, Que
| | - Caroline Lamarche
- Centre de recherche de l'Hôpital du Sacré-Cœur de Montréal (Goupil); Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM) (Benlarbi, Chatterjee, Goyette, Finzi, Suri); Centre de recherche de l'Hôpital Maisonneuve-Rosemont (Nadeau-Fredette, Lamarche), Montréal, Que.; Department of Microbiology and Immunology and Division of Nephrology, Department of Medicine (Gunaratnam), Western University, London, Ont.; Research Institute of the McGill University Health Centre (Tom, Suri); Département de microbiologie, infectiologie et immunologie (Finzi), Université de Montréal; Department of Microbiology and Immunology (Finzi), McGill University; Division of Nephrology (Suri), Department of Medicine, McGill University, Montréal, Que
| | - Alexander Tom
- Centre de recherche de l'Hôpital du Sacré-Cœur de Montréal (Goupil); Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM) (Benlarbi, Chatterjee, Goyette, Finzi, Suri); Centre de recherche de l'Hôpital Maisonneuve-Rosemont (Nadeau-Fredette, Lamarche), Montréal, Que.; Department of Microbiology and Immunology and Division of Nephrology, Department of Medicine (Gunaratnam), Western University, London, Ont.; Research Institute of the McGill University Health Centre (Tom, Suri); Département de microbiologie, infectiologie et immunologie (Finzi), Université de Montréal; Department of Microbiology and Immunology (Finzi), McGill University; Division of Nephrology (Suri), Department of Medicine, McGill University, Montréal, Que
| | - Andrés Finzi
- Centre de recherche de l'Hôpital du Sacré-Cœur de Montréal (Goupil); Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM) (Benlarbi, Chatterjee, Goyette, Finzi, Suri); Centre de recherche de l'Hôpital Maisonneuve-Rosemont (Nadeau-Fredette, Lamarche), Montréal, Que.; Department of Microbiology and Immunology and Division of Nephrology, Department of Medicine (Gunaratnam), Western University, London, Ont.; Research Institute of the McGill University Health Centre (Tom, Suri); Département de microbiologie, infectiologie et immunologie (Finzi), Université de Montréal; Department of Microbiology and Immunology (Finzi), McGill University; Division of Nephrology (Suri), Department of Medicine, McGill University, Montréal, Que
| | - Rita S Suri
- Centre de recherche de l'Hôpital du Sacré-Cœur de Montréal (Goupil); Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM) (Benlarbi, Chatterjee, Goyette, Finzi, Suri); Centre de recherche de l'Hôpital Maisonneuve-Rosemont (Nadeau-Fredette, Lamarche), Montréal, Que.; Department of Microbiology and Immunology and Division of Nephrology, Department of Medicine (Gunaratnam), Western University, London, Ont.; Research Institute of the McGill University Health Centre (Tom, Suri); Département de microbiologie, infectiologie et immunologie (Finzi), Université de Montréal; Department of Microbiology and Immunology (Finzi), McGill University; Division of Nephrology (Suri), Department of Medicine, McGill University, Montréal, Que.
| | | |
Collapse
|
656
|
Dispinseri S, Secchi M, Pirillo MF, Tolazzi M, Borghi M, Brigatti C, De Angelis ML, Baratella M, Bazzigaluppi E, Venturi G, Sironi F, Canitano A, Marzinotto I, Tresoldi C, Ciceri F, Piemonti L, Negri D, Cara A, Lampasona V, Scarlatti G. Neutralizing antibody responses to SARS-CoV-2 in symptomatic COVID-19 is persistent and critical for survival. Nat Commun 2021; 12:2670. [PMID: 33976165 PMCID: PMC8113594 DOI: 10.1038/s41467-021-22958-8] [Citation(s) in RCA: 271] [Impact Index Per Article: 67.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/11/2021] [Indexed: 12/11/2022] Open
Abstract
Understanding how antibody responses to SARS-CoV-2 evolve during infection may provide important insight into therapeutic approaches and vaccination for COVID-19. Here we profile the antibody responses of 162 COVID-19 symptomatic patients in the COVID-BioB cohort followed longitudinally for up to eight months from symptom onset to find SARS-CoV-2 neutralization, as well as antibodies either recognizing SARS-CoV-2 spike antigens and nucleoprotein, or specific for S2 antigen of seasonal beta-coronaviruses and hemagglutinin of the H1N1 flu virus. The presence of neutralizing antibodies within the first weeks from symptoms onset correlates with time to a negative swab result (p = 0.002), while the lack of neutralizing capacity correlates with an increased risk of a fatal outcome (p = 0.008). Neutralizing antibody titers progressively drop after 5-8 weeks but are still detectable up to 8 months in the majority of recovered patients regardless of age or co-morbidities, with IgG to spike antigens providing the best correlate of neutralization. Antibody responses to seasonal coronaviruses are temporarily boosted, and parallel those to SARS-CoV-2 without dampening the specific response or worsening disease progression. Our results thus suggest compromised immune responses to the SARS-CoV-2 spike to be a major trait of COVID-19 patients with critical conditions, and thereby inform on the planning of COVID-19 patient care and therapy prioritization.
Collapse
Affiliation(s)
- Stefania Dispinseri
- Viral Evolution and Transmission Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Massimiliano Secchi
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- DNA Enzymology & Molecular Virology Unit, Institute of Molecular Genetics, National Research Council, Pavia, Italy
| | | | - Monica Tolazzi
- Viral Evolution and Transmission Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Martina Borghi
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Cristina Brigatti
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Maria Laura De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Marco Baratella
- Viral Evolution and Transmission Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Giulietta Venturi
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Sironi
- Viral Evolution and Transmission Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Andrea Canitano
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Ilaria Marzinotto
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Cristina Tresoldi
- Molecular Hematology Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Fabio Ciceri
- Hematology and Bone Marrow Transplantation Unit, IRCCS Ospedale San Raffaele, Milan, Italy
- School of Medicine and Surgery, Università Vita-Salute San Raffaele, Milan, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- School of Medicine and Surgery, Università Vita-Salute San Raffaele, Milan, Italy
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Andrea Cara
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Vito Lampasona
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Gabriella Scarlatti
- Viral Evolution and Transmission Unit, IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
657
|
López-Mendoza H, Montañés A, Moliner-Lahoz FJ. Disparities in the Evolution of the COVID-19 Pandemic between Spanish Provinces. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:5085. [PMID: 34064938 PMCID: PMC8151898 DOI: 10.3390/ijerph18105085] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/04/2021] [Accepted: 05/09/2021] [Indexed: 12/11/2022]
Abstract
Spain experienced a second wave of the COVID-19 pandemic in autumn 2020, which has been approached with different measures by regional authorities. We analyze the presence of convergence in the cumulative incidence for 14 days (CI14) in provinces and self-governing cities. The Phillips-Sul methodology was used to study the grouping of behavior between provinces, and an ordered logit model was estimated to understand the forces that drive creating the different convergence clubs. We reject the presence of a single pattern of behavior in the evolution of the CI14 across territories. Four statistically different convergence clubs and an additional province (Madrid) with divergent behavior are observed. Provinces with developed agricultural and industrial economic sectors, high mobility, and a high proportion of Central and South American immigrants had the highest level of CI14. We show that the transmission of the virus is not homogeneous in the Spanish national territory. Our results are helpful for identifying differences in determinants that could explain the pandemic's evolution and for formulating hypotheses about the effectiveness of implemented measures.
Collapse
Affiliation(s)
- Héctor López-Mendoza
- Directorate-General of Public Health, Aragon Department of Health, 50017 Zaragoza, Spain;
- Preventive Medicine and Public Health Department, Lozano Blesa University Hospital, 50009 Zaragoza, Spain;
| | - Antonio Montañés
- Economic Analysis Department, University of Zaragoza, 50005 Zaragoza, Spain
| | - F. Javier Moliner-Lahoz
- Preventive Medicine and Public Health Department, Lozano Blesa University Hospital, 50009 Zaragoza, Spain;
| |
Collapse
|
658
|
Gangaev A, Ketelaars SLC, Isaeva OI, Patiwael S, Dopler A, Hoefakker K, De Biasi S, Gibellini L, Mussini C, Guaraldi G, Girardis M, Ormeno CMPT, Hekking PJM, Lardy NM, Toebes M, Balderas R, Schumacher TN, Ovaa H, Cossarizza A, Kvistborg P. Identification and characterization of a SARS-CoV-2 specific CD8 + T cell response with immunodominant features. Nat Commun 2021; 12:2593. [PMID: 33972535 PMCID: PMC8110804 DOI: 10.1038/s41467-021-22811-y] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 03/26/2021] [Indexed: 02/03/2023] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 is a continuous challenge worldwide, and there is an urgent need to map the landscape of immunogenic and immunodominant epitopes recognized by CD8+ T cells. Here, we analyze samples from 31 patients with COVID-19 for CD8+ T cell recognition of 500 peptide-HLA class I complexes, restricted by 10 common HLA alleles. We identify 18 CD8+ T cell recognized SARS-CoV-2 epitopes, including an epitope with immunodominant features derived from ORF1ab and restricted by HLA-A*01:01. In-depth characterization of SARS-CoV-2-specific CD8+ T cell responses of patients with acute critical and severe disease reveals high expression of NKG2A, lack of cytokine production and a gene expression profile inhibiting T cell re-activation and migration while sustaining survival. SARS-CoV-2-specific CD8+ T cell responses are detectable up to 5 months after recovery from critical and severe disease, and these responses convert from dysfunctional effector to functional memory CD8+ T cells during convalescence.
Collapse
Affiliation(s)
- Anastasia Gangaev
- grid.430814.aDivision of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, North Holland The Netherlands
| | - Steven L. C. Ketelaars
- grid.430814.aDivision of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, North Holland The Netherlands
| | - Olga I. Isaeva
- grid.430814.aDivision of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, North Holland The Netherlands
| | - Sanne Patiwael
- grid.430814.aDivision of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, North Holland The Netherlands
| | - Anna Dopler
- grid.430814.aDivision of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, North Holland The Netherlands
| | - Kelly Hoefakker
- grid.430814.aDivision of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, North Holland The Netherlands
| | - Sara De Biasi
- grid.7548.e0000000121697570University of Modena and Reggio Emilia School of Medicine, Modena, Emilia Romagna Italy
| | - Lara Gibellini
- grid.7548.e0000000121697570University of Modena and Reggio Emilia School of Medicine, Modena, Emilia Romagna Italy
| | - Cristina Mussini
- grid.7548.e0000000121697570University of Modena and Reggio Emilia School of Medicine, Modena, Emilia Romagna Italy
| | - Giovanni Guaraldi
- grid.7548.e0000000121697570University of Modena and Reggio Emilia School of Medicine, Modena, Emilia Romagna Italy
| | - Massimo Girardis
- grid.7548.e0000000121697570University of Modena and Reggio Emilia School of Medicine, Modena, Emilia Romagna Italy
| | - Cami M. P. Talavera Ormeno
- grid.10419.3d0000000089452978Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, South Holland The Netherlands
| | - Paul J. M. Hekking
- grid.10419.3d0000000089452978Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, South Holland The Netherlands
| | - Neubury M. Lardy
- grid.417732.40000 0001 2234 6887Department of Immunogenetics, Sanquin Diagnostics B.V., Amsterdam, North Holland The Netherlands
| | - Mireille Toebes
- grid.430814.aDivision of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, North Holland The Netherlands
| | - Robert Balderas
- grid.420052.10000 0004 0543 6807Department of Biological Sciences, BD Biosciences, San Jose, CA USA
| | - Ton N. Schumacher
- grid.430814.aDivision of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, North Holland The Netherlands
| | - Huib Ovaa
- grid.10419.3d0000000089452978Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, South Holland The Netherlands
| | - Andrea Cossarizza
- grid.7548.e0000000121697570University of Modena and Reggio Emilia School of Medicine, Modena, Emilia Romagna Italy
| | - Pia Kvistborg
- grid.430814.aDivision of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, North Holland The Netherlands
| |
Collapse
|
659
|
Brustolin M, Rodon J, Rodríguez de la Concepción ML, Ávila-Nieto C, Cantero G, Pérez M, Te N, Noguera-Julián M, Guallar V, Valencia A, Roca N, Izquierdo-Useros N, Blanco J, Clotet B, Bensaid A, Carrillo J, Vergara-Alert J, Segalés J. Protection against reinfection with D614- or G614-SARS-CoV-2 isolates in golden Syrian hamster. Emerg Microbes Infect 2021; 10:797-809. [PMID: 33825619 PMCID: PMC8812745 DOI: 10.1080/22221751.2021.1913974] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Reinfections with SARS-CoV-2 have already been documented in humans, although its real incidence is currently unknown. Besides having a great impact on public health, this phenomenon raises the question of immunity generated by a single infection is sufficient to provide sterilizing/protective immunity to a subsequent SARS-CoV-2 re-exposure. The Golden Syrian hamster is a manageable animal model to explore immunological mechanisms able to counteract COVID-19, as it recapitulates pathological aspects of mild to moderately affected patients. Here, we report that SARS-CoV-2-inoculated hamsters resolve infection in the upper and lower respiratory tracts within seven days upon inoculation with the Cat01 (G614) SARS-CoV-2 isolate. Three weeks after the primary challenge, and despite high titres of neutralizing antibodies, half of the animals were susceptible to reinfection by both identical (Cat01, G614) and variant (WA/1, D614) SARS-CoV-2 isolates. However, upon re-inoculation, only nasal tissues were transiently infected with much lower viral replication than those observed after the first inoculation. These data indicate that a primary SARS-CoV-2 infection is not sufficient to elicit a sterilizing immunity in hamster models but protects against lung disease.
Collapse
Affiliation(s)
- Marco Brustolin
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Caldes de Montbui, Spain
| | - Jordi Rodon
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Caldes de Montbui, Spain
| | | | | | - Guillermo Cantero
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Caldes de Montbui, Spain
| | - Mónica Pérez
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Caldes de Montbui, Spain
| | - Nigeer Te
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Caldes de Montbui, Spain
| | - Marc Noguera-Julián
- IrsiCaixa AIDS Research Institute, Badalona, Spain.,University of Vic-Central University of Catalonia (UVic-UCC), Catalonia, Spain
| | - Víctor Guallar
- Barcelona Supercomputing Center (BSC), Barcelona, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Alfonso Valencia
- Barcelona Supercomputing Center (BSC), Barcelona, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Núria Roca
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Caldes de Montbui, Spain
| | - Nuria Izquierdo-Useros
- IrsiCaixa AIDS Research Institute, Badalona, Spain.,Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | - Julià Blanco
- IrsiCaixa AIDS Research Institute, Badalona, Spain.,University of Vic-Central University of Catalonia (UVic-UCC), Catalonia, Spain.,Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute, Badalona, Spain.,University of Vic-Central University of Catalonia (UVic-UCC), Catalonia, Spain.,Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | - Albert Bensaid
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Caldes de Montbui, Spain
| | | | - Júlia Vergara-Alert
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Caldes de Montbui, Spain
| | - Joaquim Segalés
- UAB, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Barcelona, Spain.,Departament de Sanitat i Anatomia Animals, Facultat de Veterinària, UAB, Barcelona, Spain
| |
Collapse
|
660
|
Abstract
The landscape of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) diagnostic testing is rapidly evolving. While serology testing has limited diagnostic capacity for acute infection, its role in providing population-based information on positivity rates and informing evidence-based decision making for public health recommendations is increasing. With the global availability of vaccines, there is increasing pressure on clinical laboratories to provide antibody screening and result interpretation for vaccinated and non-vaccinated individuals. Here we present the most up-to-date data on SARS-CoV-2 antibody timelines, including the longevity of antibodies, and the production and detection of neutralizing antibodies. Additionally, we provide practical guidance for clinical microbiology laboratories to both verify commercial serology assays and choose appropriate testing algorithms for their local populations.
Collapse
|
661
|
Sonnleitner ST, Prelog M, Jansen B, Rodgarkia-Dara C, Gietl S, Schönegger CM, Koblmüller S, Sturmbauer C, Posch W, Almanzar G, Jury H, Loney T, Tichy A, Nowotny N, Walder G. Maintenance of neutralizing antibodies over ten months in convalescent SARS-CoV-2 afflicted patients. Transbound Emerg Dis 2021; 69:1596-1605. [PMID: 33960696 PMCID: PMC8242897 DOI: 10.1111/tbed.14130] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/30/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022]
Abstract
Knowledge of the level and duration of protective immunity against SARS‐CoV‐2 after primary infection is of crucial importance for preventive approaches. Currently, there is a lack of evidence on the persistence of specific antibodies. We investigated the generation and maintenance of neutralizing antibodies of convalescent SARS‐CoV‐2‐afflicted patients over a ten‐month period post‐primary infection using an immunofluorescence assay, a commercial chemiluminescent immunoassay and an in‐house enzyme‐linked neutralization assay. We present the successful application of an improved version of the plaque‐reduction neutralization assay which can be analysed optometrically to simplify data interpretation. Based on the results of the enzyme‐linked neutralization assay, neutralizing antibodies were maintained in 77.4% of convalescent individuals without relevant decay over ten months. Furthermore, a positive correlation between severity of infection and antibody titre was observed. In conclusion, SARS‐CoV‐2‐afflicted individuals have been proven to be able to develop and maintain neutralizing antibodies over a period of ten months after primary infection. Findings suggest long‐lasting presumably protective humoral immune responses after wild‐type infection.
Collapse
Affiliation(s)
- Sissy Therese Sonnleitner
- Department of Virology, Medical Laboratory, Dr. Gernot Walder GmbH, Ausservillgraten, Austria.,Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Martina Prelog
- Department of Pediatrics, Pediatric Rheumatology/Special Immunology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Bianca Jansen
- Department of Virology, Medical Laboratory, Dr. Gernot Walder GmbH, Ausservillgraten, Austria
| | | | - Sarah Gietl
- Department of Virology, Medical Laboratory, Dr. Gernot Walder GmbH, Ausservillgraten, Austria
| | - Carmen Maria Schönegger
- Department of Virology, Medical Laboratory, Dr. Gernot Walder GmbH, Ausservillgraten, Austria
| | | | | | - Wilfried Posch
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Giovanni Almanzar
- Department of Pediatrics, Pediatric Rheumatology/Special Immunology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Hanna Jury
- Department of Pediatrics, Pediatric Rheumatology/Special Immunology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Tom Loney
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Alexander Tichy
- Department for Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Norbert Nowotny
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates.,Viral Zoonoses, Emerging and Vector-Borne Infections Group, Institute of Virology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Gernot Walder
- Department of Virology, Medical Laboratory, Dr. Gernot Walder GmbH, Ausservillgraten, Austria
| |
Collapse
|
662
|
Goto A, Go H, Miyakawa K, Yamaoka Y, Ohtake N, Kubo S, Jeremiah SS, Mihara T, Senuki K, Miyazaki T, Ikeda S, Ogura T, Kato H, Matsuba I, Sanno N, Miyakawa M, Ozaki H, Kikuoka M, Ohashi Y, Ryo A, Yamanaka T. Sustained Neutralizing Antibodies 6 Months Following Infection in 376 Japanese COVID-19 Survivors. Front Microbiol 2021; 12:661187. [PMID: 34025615 PMCID: PMC8137897 DOI: 10.3389/fmicb.2021.661187] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/09/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: There is scarce evidence regarding the long-term persistence of neutralizing antibodies among coronavirus disease 2019 (COVID-19) survivors. This study determined neutralizing antibody titers (NT50) and antibodies against spike protein (SP) or nucleocapsid protein (NP) antigens approximately 6 months after the diagnosis of COVID-19. Methods: COVID-19 survivors in Japan were recruited. Serum samples and data related to patients' characteristics and COVID-19 history were collected. NT50 and titers of antibodies against NP and SP antigens were measured at 20-32 weeks after the first positive severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) test results. Factors associated with NT50 were identified using the multivariable linear regression and the correlations among NT50 and titers of immunoglobulin G (IgG) and total immunoglobulins (Igs) against NP and SP were assessed by Spearman's correlation. Results: Among 376 participants (median [range] days after testing positive for SARS-CoV-2, 180 (147-224); median [range] years of age, 50 (20-78); 188 [50%] male), most tested positive for NT50 (n = 367, 98%), SP-IgG (n = 344, 91%), SP-total Ig (n = 369, 98%), NP-IgG (n = 314, 84%), and NP-total Ig (n = 365, 97%). Regression analysis indicated that higher BMI, fever, and the requirement of mechanical ventilation or extracorporeal membrane oxygenation were significantly associated with higher NT50. Anti-SP antibodies correlated moderately with NT50 (Spearman's correlation: 0.63 for SP IgG; 0.57 for SP-total Ig), while the correlation was weak for anti-NP antibodies (0.37 for NP IgG; 0.32 for NP-total Ig). Conclusions: Most COVID-19 survivors had sustained neutralizing antibodies and tested positive for SP-total Ig and NP-total Ig approximately 6 months after infection.
Collapse
Affiliation(s)
- Atsushi Goto
- Department of Health Data Science, Yokohama City University Graduate School of Data Science, Yokohama, Japan
| | - Hirofumi Go
- Department of Biostatistics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kei Miyakawa
- Department of Microbiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yutaro Yamaoka
- Department of Microbiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Life Science Laboratory, Technology and Development Division, Kanto Chemical Co, Inc., Kanagawa, Japan
| | - Norihisa Ohtake
- Bioscience Division, Research and Development Department, Tosoh Corporation, Tokyo Research Center, Kanagawa, Japan
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Sousuke Kubo
- Department of Microbiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | | | - Takahiro Mihara
- Department of Health Data Science, Yokohama City University Graduate School of Data Science, Yokohama, Japan
| | - Kotaro Senuki
- YCU Center for Novel and Exploratory Clinical Trials, Yokohama City University Hospital, Yokohama, Japan
| | - Tomoyuki Miyazaki
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Satoshi Ikeda
- Department of Respiratory Medicine, Kanagawa Cardiovascular and Respiratory Center, Yokohama, Japan
| | - Takashi Ogura
- Department of Respiratory Medicine, Kanagawa Cardiovascular and Respiratory Center, Yokohama, Japan
| | - Hideaki Kato
- Infection Prevention and Control Department, Yokohama City University Hospital, Yokohama, Japan
| | | | | | | | | | | | - Yasuo Ohashi
- Department of Integrated Science and Engineering for Sustainable Society, Chuo University, Tokyo, Japan
| | - Akihide Ryo
- Department of Microbiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takeharu Yamanaka
- Department of Health Data Science, Yokohama City University Graduate School of Data Science, Yokohama, Japan
- Department of Biostatistics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
663
|
Affiliation(s)
- Benjamin Meyer
- Centre for Vaccinology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| |
Collapse
|
664
|
Sakhi H, Dahmane D, Attias P, Kofman T, Bouvier M, Lapidus N, Fourati S, El Karoui K. Kinetics of Anti-SARS-CoV-2 IgG Antibodies in Hemodialysis Patients Six Months after Infection. J Am Soc Nephrol 2021; 32:1033-1036. [PMID: 33637518 PMCID: PMC8259690 DOI: 10.1681/asn.2020111618] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/24/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND The humoral response against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the hemodialysis population, including its dynamics over time, remains poorly understood. METHODS To analyze initial and long-term humoral responses against SARS-CoV-2 in a hemodialysis population, we retrospectively evaluated findings from SARS-CoV-2 IgG serologic assays targeting the nucleocapsid antigen or spike antigen up to 6 months of follow-up in patients on hemodialysis in the Paris, France, region who had recovered from coronavirus disease 2019 (COVID-19). RESULTS Our analysis included 83 patients (median age 65 years); 59 (71%) were male and 28 (34%) had presented with severe COVID-19. We observed positive initial SARS-CoV-2 IgG antinucleocapsid serology in 74 patients (89%) at a median of 67 days postdiagnosis. By multivariable analysis, immunocompromised status was the only factor significantly associated with lack of an IgG antinucleocapsid antibody response. Follow-up data were available at 6 months postdiagnosis for 60 of 74 patients (81%) with positive initial antinucleocapsid serology, and 15 (25%) of them had negative antinucleocapsid serology at month 6. In total, 14 of 15 sera were tested for antispike antibodies, 3 of 14 (21%) of which were also negative. Overall, 97% of antinucleocapsid-antibody-positive specimens were also antispike-antibody positive. Female sex, age >70 years, and nonsevere clinical presentation were independently associated with faster IgG antinucleocapsid titer decay in multivariable analysis. After adjustment for sex and age >70 years, nonsevere clinical presentation was the only factor associated with faster decay of IgG antispike antibodies. CONCLUSIONS This study characterizes evolution of the SARS-CoV-2 antibody response in patients on hemodialysis and identifies factors that are associated with lack of seroconversion and with IgG titer decay.
Collapse
Affiliation(s)
- Hamza Sakhi
- Assistance Publique des Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Department of Nephrology, Centre de Référence Maladie Rare « Syndrome Néphrotique Idiopathique », Fédération Hospitalo-Universitaire « Innovative therapy for immune disorders », Créteil, France,University Paris Est Créteil, Institut National de la Santé et de la Recherche Médical, (INSERM) U955, Institut Mondor de Recherche Biomédicale (IMRB), Equipe 21, Créteil, France
| | - Djamal Dahmane
- Assistance Publique des Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Department of Nephrology, Centre de Référence Maladie Rare « Syndrome Néphrotique Idiopathique », Fédération Hospitalo-Universitaire « Innovative therapy for immune disorders », Créteil, France
| | - Philippe Attias
- Department of Nephrology and Dialysis, Hôpital Privé Nord Parisien, Sarcelles, France
| | - Thomas Kofman
- Association Néphrologique Développement Rein Artificiel, Unité de Dialyse des Buttes Chaumont, Paris, France
| | - Magali Bouvier
- Assistance Publique des Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Service de Virologie, Centre National de Référence Hépatites B, C, et delta, Créteil, France.,Department of Virology, University Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale, Créteil, France
| | - Nathanael Lapidus
- Public Health Department, Saint-Antoine Hospital, Paris, France,Sorbonne Université, Institut Pierre Louis d’Epidémiologie et de Santé Publique, Paris, France
| | - Slim Fourati
- Assistance Publique des Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Service de Virologie, Centre National de Référence Hépatites B, C, et delta, Créteil, France.,Department of Virology, University Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale, Créteil, France
| | - Khalil El Karoui
- Assistance Publique des Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Department of Nephrology, Centre de Référence Maladie Rare « Syndrome Néphrotique Idiopathique », Fédération Hospitalo-Universitaire « Innovative therapy for immune disorders », Créteil, France,University Paris Est Créteil, Institut National de la Santé et de la Recherche Médical, (INSERM) U955, Institut Mondor de Recherche Biomédicale (IMRB), Equipe 21, Créteil, France
| | | |
Collapse
|
665
|
Gallagher KM, Leick MB, Larson RC, Berger TR, Katsis K, Yam JY, Brini G, Grauwet K, Maus MV. SARS -CoV-2 T-cell immunity to variants of concern following vaccination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.05.03.442455. [PMID: 33972942 PMCID: PMC8109204 DOI: 10.1101/2021.05.03.442455] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recently, two mRNA vaccines to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have become available, but there is also an emergence of SARS-CoV-2 variants with increased transmissibility and virulence1-6. A major concern is whether the available vaccines will be equally effective against these variants. The vaccines are designed to induce an immune response against the SARS-CoV-2 spike protein7,8, which is required for viral entry to host cells9. Immunity to SARS-CoV-2 is often evaluated by antibody production, while less is known about the T-cell response. Here we developed, characterized, and implemented two standardized, functional assays to measure T-cell immunity to SARS-CoV-2 in uninfected, convalescent, and vaccinated individuals. We found that vaccinated individuals had robust T-cell responses to the wild type spike and nucleocapsid proteins, even more so than convalescent patients. We also found detectable but diminished T-cell responses to spike variants (B.1.1.7, B.1.351, and B.1.1.248) among vaccinated but otherwise healthy donors. Since decreases in antibody neutralization have also been observed with some variants10-12, investigation into the T-cell response to these variants as an alternative means of viral control is imperative. Standardized measurements of T-cell responses to SARS-CoV-2 are feasible and can be easily adjusted to determine changes in response to variants.
Collapse
Affiliation(s)
- Kathleen M.E. Gallagher
- Immune Monitoring Laboratory, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | - Mark B. Leick
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Rebecca C. Larson
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Trisha R. Berger
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Katelin Katsis
- Immune Monitoring Laboratory, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jennifer Y. Yam
- Immune Monitoring Laboratory, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Gabrielle Brini
- Immune Monitoring Laboratory, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Korneel Grauwet
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Marcela V. Maus
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
666
|
Kedl RM. Down but far from out: The durability of SARS-CoV-2 immunity after asymptomatic infection. J Exp Med 2021; 218:212001. [PMID: 33861846 PMCID: PMC8056771 DOI: 10.1084/jem.20210359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The dynamics of immune responses in asymptomatic SARS-CoV-2–infected subjects remain to be fully characterized. The work presented in this issue of JEM by Le Bert et al. (2021. J. Exp. Med.https://doi.org/10.1084/jem.20202617) sheds some light on these issues and ultimately provides some degree of confidence in the magnitude and persistence of immunity over time after asymptomatic infection with SARS-CoV-2.
Collapse
Affiliation(s)
- Ross M Kedl
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, Aurora, CO
| |
Collapse
|
667
|
Barrios Y, Franco A, Sanchez-Machin I, Poza-Guedes P, Gonzalez-Perez R, Matheu V. A novel application of delayed-type hipersensitivity reaction to measure cellular immune response in SARS-CoV-2 exposed individuals. Clin Immunol 2021; 226:108730. [PMID: 33865990 PMCID: PMC8049849 DOI: 10.1016/j.clim.2021.108730] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 01/04/2023]
Abstract
OBJECTIVE To understand the anti-virus adaptive immune response occurring during SARS-Cov-2 infection is necessary to have methods to investigate cellular and humoral components. The goal of this study has been to investigate the utility of a specific spike-DTH test using a coronavirus recombinant protein in COVID-19 patients. METHODS DTH studies were performed by intradermal injection of a commercial recombinant spike protein from SARS-CoV-2 along with conventional serology studies. RESULTS Fifty-one COVID-19 patients were studied showing 84,3% of concordance with spike-DTH and anti-RBD-IgG. Spike-DTH was superior to identify seven more COVID-19 individuals. A high specificity was found with no positive spike DTH reactions in the non-sick individuals. The skin test also showed more stable results over time while specific anti-RBD-IgG decreased gradually. Clinical severity groups also showed a progressive gradient of larger positive spike-DTH. CONCLUSION Specific spike DTH test seems to be an easy method to study cell immune response.
Collapse
Affiliation(s)
- Yvelise Barrios
- Immunology Lab, Central Lab, Floor - 1, Main Building, Hospital Universitario de Canarias, Ctra Ofra s/n. La Cuesta, 38320 La Laguna, Tenerife, Spain
| | - Andres Franco
- Immunology Lab, Central Lab, Floor - 1, Main Building, Hospital Universitario de Canarias, Ctra Ofra s/n. La Cuesta, 38320 La Laguna, Tenerife, Spain
| | - Inmaculada Sanchez-Machin
- Allergy Service, Floor-2, Outpatient Building, Hospital Universitario de Canarias, Ctra Ofra s/n. La Cuesta, 38320 La Laguna, Tenerife, Spain
| | - Paloma Poza-Guedes
- Allergy Service, Floor-2, Outpatient Building, Hospital Universitario de Canarias, Ctra Ofra s/n. La Cuesta, 38320 La Laguna, Tenerife, Spain
| | - Ruperto Gonzalez-Perez
- Allergy Service, Floor-2, Outpatient Building, Hospital Universitario de Canarias, Ctra Ofra s/n. La Cuesta, 38320 La Laguna, Tenerife, Spain
| | - Victor Matheu
- Allergy Service, Floor-2, Outpatient Building, Hospital Universitario de Canarias, Ctra Ofra s/n. La Cuesta, 38320 La Laguna, Tenerife, Spain,Corresponding author
| |
Collapse
|
668
|
Wang Z, Yang L, Chen Y, Xu Z, Wang H, Zhang X. A longitudinal follow-up of COVID-19 patients in the convalescent phase showed recovery in radiological results, the dynamics of lymphocytes, and a decrease in the level of IgG antibody: a single-centre, observational study. J Thorac Dis 2021; 13:2986-3000. [PMID: 34164190 PMCID: PMC8182512 DOI: 10.21037/jtd-20-3011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Background Given the high prevalence of coronavirus disease 2019 (COVID-19) globally, and the increased number of patients being discharged, it is necessary to understand the health consequences of COVID-19 to formulate and manage public policy concerning convalescent patients. Methods A longitudinal follow-up investigation of 25 patients from a tertiary hospital in Henan provincial was conducted 8 weeks after discharge. Of these patients, 15 attended a second follow-up appointment 8 weeks after that. A throat swab reverse transcription-polymerase chain reaction (RT-PCR) analysis for SARS-CoV-2 and chest computerized tomography (CT) scans were implemented at the first follow-up appointment, and a total of 40 blood samples (25 from the first and 15 from the second follow-up appointment) were collected. Patients’ levels of Immunoglobulin G (IgG) antibody against S-Receptor binding domain (S-RBD) and Nucleocapsid Protein (NP) of SARS-CoV-2 and the subpopulation of lymphocytes were evaluated using an enzyme-linked immunoassay (ELISA) test and flow cytometry, respectively. Results At the first follow-up appointment, 10 of the 25 patients (40.0%) showed complete radiological resolution. Of these patients, 80.0% were classified as moderate, and 80.0% were younger (those whose age was ≤ the median age of all the patients). The predominant patterns of abnormalities included an irregular line (12/25, 48.0%), ground-glass opacity (GGO) (44.0%), and multiple GGOs (28.0%). At the first follow-up appointment, 40.0% (10/25) of patients still had lymphopenia. Of the 15 patients who were followed-up with twice, the ratio of lymphopenia was 80% (12/15), 60.0% (9/15), and 46.7% (7/15) at 0, 8, and 16 weeks after discharge, respectively. This was mainly due to a decrease in the cluster of differentiation (CD) 4+ T lymphocyte, which was observed in 60% (9/15), 60% (9/15), and 46.7% (7/15) of total patients at 0, 8, and 16 weeks after discharge, respectively. All of the patients were S-RBD and NP IgG antibody positive at the first follow-up appointment. 40.0% (6/15) and 66.7% (10/15) of patients showed a decrease over 50.0% in the level of NP and S-RBD IgG antibodies, respectively, at the second follow-up appointment. The NP and S-RBD IgG antibodies’ levels declined to 44.6% (P=0.044) and 28.1% (P=0.18), respectively. 0 and 26.7% (4/15) of patients turned from NP and S-RBD IgG antibody positive to negative, respectively. Conclusions About half of the patients still showed at least 1 abnormality in chest CT scans 8 weeks after discharge and lymphopenia 16 weeks after discharge. The level of the IgG antibody had declined by the follow-up appointment. Notably, the S-RBD IgG antibody declined more dramatically than that of NP. These results may have implications in the making of policies related to disease prevention, the long-term management of discharged patients, and vaccines’ development.
Collapse
Affiliation(s)
- Ziqi Wang
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Li Yang
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Yi Chen
- Clinical Research Service Centre, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
| | - Zhiwei Xu
- Clinical Research Service Centre, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
| | - Hui Wang
- Xinxiang Medical School, Xinxiang, China.,Henan Provincial People's Hospital, Zhengzhou, China
| | - Xiaoju Zhang
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| |
Collapse
|
669
|
Zuo J, Dowell AC, Pearce H, Verma K, Long HM, Begum J, Aiano F, Amin-Chowdhury Z, Hoschler K, Brooks T, Taylor S, Hewson J, Hallis B, Stapley L, Borrow R, Linley E, Ahmad S, Parker B, Horsley A, Amirthalingam G, Brown K, Ramsay ME, Ladhani S, Moss P. Robust SARS-CoV-2-specific T cell immunity is maintained at 6 months following primary infection. Nat Immunol 2021; 22:620-626. [PMID: 33674800 PMCID: PMC7610739 DOI: 10.1038/s41590-021-00902-8] [Citation(s) in RCA: 266] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 02/19/2021] [Indexed: 12/21/2022]
Abstract
The immune response to SARS-CoV-2 is critical in controlling disease, but there is concern that waning immunity may predispose to reinfection. We analyzed the magnitude and phenotype of the SARS-CoV-2-specific T cell response in 100 donors at 6 months following infection. T cell responses were present by ELISPOT and/or intracellular cytokine staining analysis in all donors and characterized by predominant CD4+ T cell responses with strong interleukin (IL)-2 cytokine expression. Median T cell responses were 50% higher in donors who had experienced a symptomatic infection, indicating that the severity of primary infection establishes a 'set point' for cellular immunity. T cell responses to spike and nucleoprotein/membrane proteins were correlated with peak antibody levels. Furthermore, higher levels of nucleoprotein-specific T cells were associated with preservation of nucleoprotein-specific antibody level although no such correlation was observed in relation to spike-specific responses. In conclusion, our data are reassuring that functional SARS-CoV-2-specific T cell responses are retained at 6 months following infection.
Collapse
Affiliation(s)
- Jianmin Zuo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Alexander C Dowell
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Hayden Pearce
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Kriti Verma
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Heather M Long
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Jusnara Begum
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Felicity Aiano
- Immunisation and Countermeasures Division, National Infection Service, London, UK
| | - Zahin Amin-Chowdhury
- Immunisation and Countermeasures Division, National Infection Service, London, UK
| | - Katja Hoschler
- Immunisation and Countermeasures Division, National Infection Service, London, UK
| | - Tim Brooks
- Immunoassay Laboratory, National Infection Service, Porton Down, UK
| | - Stephen Taylor
- Immunoassay Laboratory, National Infection Service, Porton Down, UK
| | - Jacqueline Hewson
- Immunisation and Countermeasures Division, National Infection Service, London, UK
| | - Bassam Hallis
- Immunoassay Laboratory, National Infection Service, Porton Down, UK
| | - Lorrain Stapley
- Immunoassay Laboratory, National Infection Service, Porton Down, UK
| | - Ray Borrow
- Sero-epidemiology Unit, Public Health England, Public Health Laboratory Manchester, Manchester Medical Microbiology Partnership, Manchester Royal Infirmary, Manchester, UK
| | - Ezra Linley
- Sero-epidemiology Unit, Public Health England, Public Health Laboratory Manchester, Manchester Medical Microbiology Partnership, Manchester Royal Infirmary, Manchester, UK
| | - Shazaad Ahmad
- Department of Virology, Manchester Medical Microbiology Partnership, Manchester Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Ben Parker
- The NIHR Manchester Clinical Research Facility, Manchester University NHS Foundation Trust, Manchester, UK
- Kellgren Centre for Rheumatology, NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, UK
| | - Alex Horsley
- The NIHR Manchester Clinical Research Facility, Manchester University NHS Foundation Trust, Manchester, UK
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester, UK
| | | | - Kevin Brown
- Immunisation and Countermeasures Division, National Infection Service, London, UK
| | - Mary E Ramsay
- Immunisation and Countermeasures Division, National Infection Service, London, UK
| | - Shamez Ladhani
- Immunisation and Countermeasures Division, National Infection Service, London, UK.
- Paediatric Infectious Diseases Research Group (PIDRG), St. Georges University of London (SGUL), London, UK.
| | - Paul Moss
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
| |
Collapse
|
670
|
Borgonovo F, Passerini M, Piscaglia M, Morena V, Giacomelli A, Oreni L, Dedivitiis G, Lupo A, Falvella S, Cossu MV, Capetti AF. Is COVID-19 severity associated with anti-spike antibody duration? Data from the ARCOVID prospective observational study. J Infect 2021; 82:e28-e30. [PMID: 33545166 PMCID: PMC7857121 DOI: 10.1016/j.jinf.2021.01.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 01/30/2021] [Indexed: 12/23/2022]
Affiliation(s)
- Fabio Borgonovo
- Department of Infectious Diseases, ASST Fatebenefratelli-Sacco, Luigi Sacco University Hospital, Milan, Italy.
| | - Matteo Passerini
- Luigi Sacco Department of Biomedical and Clinical Sciences DIBIC, University of Milan, Italy
| | - Marco Piscaglia
- Luigi Sacco Department of Biomedical and Clinical Sciences DIBIC, University of Milan, Italy
| | - Valentina Morena
- Luigi Sacco Department of Biomedical and Clinical Sciences DIBIC, University of Milan, Italy
| | - Andrea Giacomelli
- Department of Infectious Diseases, ASST Fatebenefratelli-Sacco, Luigi Sacco University Hospital, Milan, Italy; Luigi Sacco Department of Biomedical and Clinical Sciences DIBIC, University of Milan, Italy
| | - Letizia Oreni
- Department of Infectious Diseases, ASST Fatebenefratelli-Sacco, Luigi Sacco University Hospital, Milan, Italy
| | - Gianfranco Dedivitiis
- Department of Infectious Diseases, ASST Fatebenefratelli-Sacco, Luigi Sacco University Hospital, Milan, Italy
| | - Angelica Lupo
- Department of Infectious Diseases, ASST Fatebenefratelli-Sacco, Luigi Sacco University Hospital, Milan, Italy; Luigi Sacco Department of Biomedical and Clinical Sciences DIBIC, University of Milan, Italy
| | | | - Maria Vittoria Cossu
- Department of Infectious Diseases, ASST Fatebenefratelli-Sacco, Luigi Sacco University Hospital, Milan, Italy
| | - Amedeo F Capetti
- Department of Infectious Diseases, ASST Fatebenefratelli-Sacco, Luigi Sacco University Hospital, Milan, Italy; Luigi Sacco Department of Biomedical and Clinical Sciences DIBIC, University of Milan, Italy
| |
Collapse
|
671
|
Kamat D, Kamat A, Mathur A. Immune Responses in Patients with COVID-19: An Overview. Pediatr Ann 2021; 50:e222-e226. [PMID: 34044706 DOI: 10.3928/19382359-20210421-02] [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] [Indexed: 11/20/2022]
Abstract
Since late 2019, the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection has resulted in more than 143 million confirmed infections and more than 3 million deaths worldwide (as of publication time). In this article, we discuss current knowledge of immune responses that confer protection to more than 80% of the people who have been infected and possible mechanisms by which the virus escapes immune surveillance in people who develop severe disease and those who die from the disease. We also discuss the differences in the immune responses by which, in most children, the infection results in only mild disease, although causing severe disease in some adults. Understanding these differences in both the innate and adaptive immune responses among these people can lead to the development of biotherapeutic treatment modalities that could modulate immune responses to offer protection against SARS-CoV-2 and block the ability of the virus to cause severe disease or death in humans. [Pediatr Ann. 2021;50(5):e222-e226.].
Collapse
|
672
|
Ward H, Cooke GS, Atchison C, Whitaker M, Elliott J, Moshe M, Brown JC, Flower B, Daunt A, Ainslie K, Ashby D, Donnelly CA, Riley S, Darzi A, Barclay W, Elliott P. Prevalence of antibody positivity to SARS-CoV-2 following the first peak of infection in England: Serial cross-sectional studies of 365,000 adults. THE LANCET REGIONAL HEALTH. EUROPE 2021; 4:100098. [PMID: 33969335 PMCID: PMC8088780 DOI: 10.1016/j.lanepe.2021.100098] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND The time-concentrated nature of the first wave of the COVID-19 epidemic in England in March and April 2020 provides a natural experiment to measure changes in antibody positivity at the population level before onset of the second wave and initiation of the vaccination programme. METHODS Three cross-sectional national surveys with non-overlapping random samples of the population in England undertaken between late June and September 2020 (REACT-2 study). 365,104 adults completed questionnaires and self-administered lateral flow immunoassay (LFIA) tests for IgG against SARS-CoV-2. FINDINGS Overall, 17,576 people had detectable antibodies, a prevalence of 4.9% (95% confidence intervals 4.9, 5.0) when adjusted for test characteristics and weighted to the adult population of England. The prevalence declined from 6.0% (5.8, 6.1), to 4.8% (4.7, 5.0) and 4.4% (4.3, 4.5), over the three rounds of the study a difference of -26.5% (-29.0, -23.8). The highest prevalence and smallest overall decline in positivity was in the youngest age group (18-24 years) at -14.9% (-21.6, -8.1), and lowest prevalence and largest decline in the oldest group (>74 years) at -39.0% (-50.8, -27.2). The decline from June to September 2020 was largest in those who did not report a history of COVID-19 at -64.0% (-75.6, -52.3), compared to -22.3% (-27.0, -17.7) in those with SARS-CoV-2 infection confirmed on PCR. INTERPRETATION A large proportion of the population remained susceptible to SARS-CoV-2 infection in England based on naturally acquired immunity from the first wave. Widespread vaccination is needed to confer immunity and control the epidemic at population level. FUNDING This work was funded by the Department of Health and Social Care in England.
Collapse
Affiliation(s)
- Helen Ward
- School of Public Health, Imperial College London, London W2 1PG, UK
- Imperial College Healthcare NHS Trust, London, UK
- MRC Centre for Global infectious Disease Analysis and Abdul Latif Jameel Institute for Disease and Emergency Analytics, Imperial College London, London, UK
| | - Graham S. Cooke
- Department of Infectious Disease, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, London, UK
| | - Christina Atchison
- School of Public Health, Imperial College London, London W2 1PG, UK
- Imperial College Healthcare NHS Trust, London, UK
| | - Matthew Whitaker
- School of Public Health, Imperial College London, London W2 1PG, UK
| | - Joshua Elliott
- School of Public Health, Imperial College London, London W2 1PG, UK
| | - Maya Moshe
- Department of Infectious Disease, Imperial College London, London, UK
| | - Jonathan C Brown
- Department of Infectious Disease, Imperial College London, London, UK
| | - Barnaby Flower
- Department of Infectious Disease, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, London, UK
| | - Anna Daunt
- Department of Infectious Disease, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, London, UK
| | - Kylie Ainslie
- School of Public Health, Imperial College London, London W2 1PG, UK
- MRC Centre for Global infectious Disease Analysis and Abdul Latif Jameel Institute for Disease and Emergency Analytics, Imperial College London, London, UK
| | - Deborah Ashby
- School of Public Health, Imperial College London, London W2 1PG, UK
| | - Christl A. Donnelly
- School of Public Health, Imperial College London, London W2 1PG, UK
- MRC Centre for Global infectious Disease Analysis and Abdul Latif Jameel Institute for Disease and Emergency Analytics, Imperial College London, London, UK
- Department of Statistics, University of Oxford, UK
| | - Steven Riley
- School of Public Health, Imperial College London, London W2 1PG, UK
- MRC Centre for Global infectious Disease Analysis and Abdul Latif Jameel Institute for Disease and Emergency Analytics, Imperial College London, London, UK
| | - Ara Darzi
- Institute for Global Health Innovation, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, London, UK
| | - Wendy Barclay
- Department of Infectious Disease, Imperial College London, London, UK
| | - Paul Elliott
- School of Public Health, Imperial College London, London W2 1PG, UK
- Imperial College Healthcare NHS Trust, London, UK
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, UK
- Health Data Research (HDR) UK London, Imperial College London, London, UK
- Dementia Research Institute, Imperial College London, London, UK
| |
Collapse
|
673
|
Harvey RA, Rassen JA, Kabelac CA, Turenne W, Leonard S, Klesh R, Meyer WA, Kaufman HW, Anderson S, Cohen O, Petkov VI, Cronin KA, Van Dyke AL, Lowy DR, Sharpless NE, Penberthy LT. Association of SARS-CoV-2 Seropositive Antibody Test With Risk of Future Infection. JAMA Intern Med 2021; 181:672-679. [PMID: 33625463 PMCID: PMC7905701 DOI: 10.1001/jamainternmed.2021.0366] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 01/28/2021] [Indexed: 01/20/2023]
Abstract
Importance Understanding the effect of serum antibodies to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on susceptibility to infection is important for identifying at-risk populations and could have implications for vaccine deployment. Objective The study purpose was to evaluate evidence of SARS-CoV-2 infection based on diagnostic nucleic acid amplification test (NAAT) among patients with positive vs negative test results for antibodies in an observational descriptive cohort study of clinical laboratory and linked claims data. Design, Setting, and Participants The study created cohorts from a deidentified data set composed of commercial laboratory tests, medical and pharmacy claims, electronic health records, and hospital chargemaster data. Patients were categorized as antibody-positive or antibody-negative according to their first SARS-CoV-2 antibody test in the database. Main Outcomes and Measures Primary end points were post-index diagnostic NAAT results, with infection defined as a positive diagnostic test post-index, measured in 30-day intervals (0-30, 31-60, 61-90, >90 days). Additional measures included demographic, geographic, and clinical characteristics at the time of the index antibody test, including recorded signs and symptoms or prior evidence of coronavirus 2019 (COVID) diagnoses or positive NAAT results and recorded comorbidities. Results The cohort included 3 257 478 unique patients with an index antibody test; 56% were female with a median (SD) age of 48 (20) years. Of these, 2 876 773 (88.3%) had a negative index antibody result, and 378 606 (11.6%) had a positive index antibody result. Patients with a negative antibody test result were older than those with a positive result (mean age 48 vs 44 years). Of index-positive patients, 18.4% converted to seronegative over the follow-up period. During the follow-up periods, the ratio (95% CI) of positive NAAT results among individuals who had a positive antibody test at index vs those with a negative antibody test at index was 2.85 (95% CI, 2.73-2.97) at 0 to 30 days, 0.67 (95% CI, 0.6-0.74) at 31 to 60 days, 0.29 (95% CI, 0.24-0.35) at 61 to 90 days, and 0.10 (95% CI, 0.05-0.19) at more than 90 days. Conclusions and Relevance In this cohort study, patients with positive antibody test results were initially more likely to have positive NAAT results, consistent with prolonged RNA shedding, but became markedly less likely to have positive NAAT results over time, suggesting that seropositivity is associated with protection from infection. The duration of protection is unknown, and protection may wane over time.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Valentina I. Petkov
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Kathy A. Cronin
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Alison L. Van Dyke
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Douglas R. Lowy
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Norman E. Sharpless
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Lynne T. Penberthy
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
674
|
Jeremijenko A, Chemaitelly H, Ayoub HH, Alishaq M, Abou-Samra AB, Al Ajmi JAAA, Al Ansari NAA, Al Kanaani Z, Al Khal A, Al Kuwari E, Al-Mohammed A, Al Molawi NHA, Al Naomi HM, Butt AA, Coyle P, El Kahlout RA, Gillani I, Kaleeckal AH, Masoodi NA, Thomas AG, Nafady-Hego H, Latif AN, Shaik RM, Younes NBM, Rahim HFA, Yassine HM, Al Kuwari MG, Al Romaihi HE, Al-Thani MH, Bertollini R, Abu-Raddad LJ. Herd Immunity against Severe Acute Respiratory Syndrome Coronavirus 2 Infection in 10 Communities, Qatar. Emerg Infect Dis 2021; 27:1343-1352. [PMID: 33900174 PMCID: PMC8084480 DOI: 10.3201/eid2705.204365] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
We investigated what proportion of the population acquired severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and whether the herd immunity threshold has been reached in 10 communities in Qatar. The study included 4,970 participants during June 21-September 9, 2020. Antibodies against SARS-CoV-2 were detected by using an electrochemiluminescence immunoassay. Seropositivity ranged from 54.9% (95% CI 50.2%-59.4%) to 83.8% (95% CI 79.1%-87.7%) across communities and showed a pooled mean of 66.1% (95% CI 61.5%-70.6%). A range of other epidemiologic measures indicated that active infection is rare, with limited if any sustainable infection transmission for clusters to occur. Only 5 infections were ever severe and 1 was critical in these young communities; infection severity rate of 0.2% (95% CI 0.1%-0.4%). Specific communities in Qatar have or nearly reached herd immunity for SARS-CoV-2 infection: 65%-70% of the population has been infected.
Collapse
|
675
|
Amanat F, Thapa M, Lei T, Sayed Ahmed SM, Adelsberg DC, Carreno JM, Strohmeier S, Schmitz AJ, Zafar S, Zhou JQ, Rijnink W, Alshammary H, Borcherding N, Reiche AG, Srivastava K, Sordillo EM, van Bakel H, Turner JS, Bajic G, Simon V, Ellebedy AH, Krammer F. The plasmablast response to SARS-CoV-2 mRNA vaccination is dominated by non-neutralizing antibodies and targets both the NTD and the RBD. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.03.07.21253098. [PMID: 33758878 PMCID: PMC7987037 DOI: 10.1101/2021.03.07.21253098] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In this study we profiled vaccine-induced polyclonal antibodies as well as plasmablast derived mAbs from individuals who received SARS-CoV-2 spike mRNA vaccine. Polyclonal antibody responses in vaccinees were robust and comparable to or exceeded those seen after natural infection. However, the ratio of binding to neutralizing antibodies after vaccination was greater than that after natural infection and, at the monoclonal level, we found that the majority of vaccine-induced antibodies did not have neutralizing activity. We also found a co-dominance of mAbs targeting the NTD and RBD of SARS-CoV-2 spike and an original antigenic-sin like backboost to seasonal human coronaviruses OC43 and HKU1. Neutralizing activity of NTD mAbs but not RBD mAbs against a clinical viral isolate carrying E484K as well as extensive changes in the NTD was abolished, suggesting that a proportion of vaccine induced RBD binding antibodies may provide substantial protection against viral variants carrying single E484K RBD mutations.
Collapse
Affiliation(s)
- Fatima Amanat
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mahima Thapa
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Tinting Lei
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Shaza M. Sayed Ahmed
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Daniel C. Adelsberg
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Juan Manuel Carreno
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Shirin Strohmeier
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Aaron J. Schmitz
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sarah Zafar
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Julian Q Zhou
- AbCellera Biologics Inc., Vancouver, BC V5Y 0A1, Canada
| | - Willemijn Rijnink
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hala Alshammary
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nicholas Borcherding
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ana Gonzalez Reiche
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Emilia Mia Sordillo
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Harm van Bakel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Jackson S. Turner
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Goran Bajic
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Viviana Simon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ali H. Ellebedy
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
676
|
Chang CC, Vlad G, Vasilescu ER, Husain SA, Liu YN, Sun WZ, Chang MF, Suciu-Foca N, Mohan S. Disparity between levels of anti-RBD IgG and anti-nucleocapsid protein IgG antibodies in COVID-19-recovered patients who received a kidney transplant. Kidney Int 2021; 100:240-241. [PMID: 33964249 PMCID: PMC8084603 DOI: 10.1016/j.kint.2021.04.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 11/23/2022]
Affiliation(s)
- Chih-Chao Chang
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA.
| | - George Vlad
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
| | - Elena-Rodica Vasilescu
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
| | - Syed A Husain
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, New York, New York, USA; Department of Epidemiology, The Columbia University Renal Epidemiology (CURE) Group, Columbia University, New York, New York, USA
| | - Ya Nan Liu
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
| | - Wei-Zen Sun
- Department of Anesthesiology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ming-Fu Chang
- Institute of Biochemistry and Molecular Biology, National Taiwan University, Taipei, Taiwan
| | - Nicole Suciu-Foca
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
| | - Sumit Mohan
- Department of Medicine, Division of Nephrology, Columbia University Irving Medical Center, New York, New York, USA; Department of Epidemiology, The Columbia University Renal Epidemiology (CURE) Group, Columbia University, New York, New York, USA
| |
Collapse
|
677
|
Hoffmann M, Arora P, Groß R, Seidel A, Hörnich BF, Hahn AS, Krüger N, Graichen L, Hofmann-Winkler H, Kempf A, Winkler MS, Schulz S, Jäck HM, Jahrsdörfer B, Schrezenmeier H, Müller M, Kleger A, Münch J, Pöhlmann S. SARS-CoV-2 variants B.1.351 and P.1 escape from neutralizing antibodies. Cell 2021; 184:2384-2393.e12. [PMID: 33794143 PMCID: PMC7980144 DOI: 10.1016/j.cell.2021.03.036] [Citation(s) in RCA: 692] [Impact Index Per Article: 173.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 02/26/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023]
Abstract
The global spread of SARS-CoV-2/COVID-19 is devastating health systems and economies worldwide. Recombinant or vaccine-induced neutralizing antibodies are used to combat the COVID-19 pandemic. However, the recently emerged SARS-CoV-2 variants B.1.1.7 (UK), B.1.351 (South Africa), and P.1 (Brazil) harbor mutations in the viral spike (S) protein that may alter virus-host cell interactions and confer resistance to inhibitors and antibodies. Here, using pseudoparticles, we show that entry of all variants into human cells is susceptible to blockade by the entry inhibitors soluble ACE2, Camostat, EK-1, and EK-1-C4. In contrast, entry of the B.1.351 and P.1 variant was partially (Casirivimab) or fully (Bamlanivimab) resistant to antibodies used for COVID-19 treatment. Moreover, entry of these variants was less efficiently inhibited by plasma from convalescent COVID-19 patients and sera from BNT162b2-vaccinated individuals. These results suggest that SARS-CoV-2 may escape neutralizing antibody responses, which has important implications for efforts to contain the pandemic.
Collapse
Affiliation(s)
- Markus Hoffmann
- Infection Biology Unit, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University Göttingen, Wilhelmsplatz 1, 37073 Göttingen, Germany.
| | - Prerna Arora
- Infection Biology Unit, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University Göttingen, Wilhelmsplatz 1, 37073 Göttingen, Germany
| | - Rüdiger Groß
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstr. 1, 89081 Ulm, Germany
| | - Alina Seidel
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstr. 1, 89081 Ulm, Germany
| | - Bojan F Hörnich
- Junior Research Group Herpesviruses - Infection Biology Unit, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany
| | - Alexander S Hahn
- Junior Research Group Herpesviruses - Infection Biology Unit, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany
| | - Nadine Krüger
- Infection Biology Unit, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany
| | - Luise Graichen
- Infection Biology Unit, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany
| | - Heike Hofmann-Winkler
- Infection Biology Unit, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany
| | - Amy Kempf
- Infection Biology Unit, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University Göttingen, Wilhelmsplatz 1, 37073 Göttingen, Germany
| | - Martin S Winkler
- Department of Anaesthesiology, University of Göttingen Medical Center, Göttingen, Georg-August University of Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Sebastian Schulz
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander University of Erlangen-Nürnberg, Glückstraße 6, 91054 Erlangen, Germany
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander University of Erlangen-Nürnberg, Glückstraße 6, 91054 Erlangen, Germany
| | - Bernd Jahrsdörfer
- Department of Transfusion Medicine, Ulm University, Helmholtzstraße 10, 89081 Ulm, Germany; Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg - Hessen and University Hospital Ulm, Helmholtzstraße 10, 89081 Ulm, Germany
| | - Hubert Schrezenmeier
- Department of Transfusion Medicine, Ulm University, Helmholtzstraße 10, 89081 Ulm, Germany; Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg - Hessen and University Hospital Ulm, Helmholtzstraße 10, 89081 Ulm, Germany
| | - Martin Müller
- Department of Internal Medicine 1, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Alexander Kleger
- Department of Internal Medicine 1, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstr. 1, 89081 Ulm, Germany; Core Facility Functional Peptidomics, Ulm University Medical Center, Meyerhofstr. 4, 89081 Ulm, Germany
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University Göttingen, Wilhelmsplatz 1, 37073 Göttingen, Germany.
| |
Collapse
|
678
|
Damluji AA, Rajan D, Haymond A, deFilippi C. Serological Testing for COVID-19 Disease: Moving the Field of Serological Surveillance Forward. J Appl Lab Med 2021; 6:584-587. [PMID: 33693726 PMCID: PMC7989295 DOI: 10.1093/jalm/jfab018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/23/2021] [Indexed: 12/30/2022]
Affiliation(s)
- Abdulla A Damluji
- Inova Center of Outcomes Research, Inova Heart and Vascular Institute, Falls Church, VA, USA
| | - Devin Rajan
- Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Amanda Haymond
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Christopher deFilippi
- Inova Center of Outcomes Research, Inova Heart and Vascular Institute, Falls Church, VA, USA
| |
Collapse
|
679
|
Zhou D, Dejnirattisai W, Supasa P, Liu C, Mentzer AJ, Ginn HM, Zhao Y, Duyvesteyn HME, Tuekprakhon A, Nutalai R, Wang B, Paesen GC, Lopez-Camacho C, Slon-Campos J, Hallis B, Coombes N, Bewley K, Charlton S, Walter TS, Skelly D, Lumley SF, Dold C, Levin R, Dong T, Pollard AJ, Knight JC, Crook D, Lambe T, Clutterbuck E, Bibi S, Flaxman A, Bittaye M, Belij-Rammerstorfer S, Gilbert S, James W, Carroll MW, Klenerman P, Barnes E, Dunachie SJ, Fry EE, Mongkolsapaya J, Ren J, Stuart DI, Screaton GR. Evidence of escape of SARS-CoV-2 variant B.1.351 from natural and vaccine-induced sera. Cell 2021; 184:2348-2361.e6. [PMID: 33730597 PMCID: PMC7901269 DOI: 10.1016/j.cell.2021.02.037] [Citation(s) in RCA: 765] [Impact Index Per Article: 191.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 12/20/2022]
Abstract
The race to produce vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) began when the first sequence was published, and this forms the basis for vaccines currently deployed globally. Independent lineages of SARS-CoV-2 have recently been reported: UK, B.1.1.7; South Africa, B.1.351; and Brazil, P.1. These variants have multiple changes in the immunodominant spike protein that facilitates viral cell entry via the angiotensin-converting enzyme-2 (ACE2) receptor. Mutations in the receptor recognition site on the spike are of great concern for their potential for immune escape. Here, we describe a structure-function analysis of B.1.351 using a large cohort of convalescent and vaccinee serum samples. The receptor-binding domain mutations provide tighter ACE2 binding and widespread escape from monoclonal antibody neutralization largely driven by E484K, although K417N and N501Y act together against some important antibody classes. In a number of cases, it would appear that convalescent and some vaccine serum offers limited protection against this variant.
Collapse
Affiliation(s)
- Daming Zhou
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, The Wellcome Centre for Human Genetics, Oxford, UK
| | - Wanwisa Dejnirattisai
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Piyada Supasa
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Chang Liu
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS), Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Alexander J Mentzer
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Helen M Ginn
- Instruct-ERIC, Oxford House, Parkway Court, John Smith Drive, Oxford, UK
| | - Yuguang Zhao
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, The Wellcome Centre for Human Genetics, Oxford, UK
| | - Helen M E Duyvesteyn
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, The Wellcome Centre for Human Genetics, Oxford, UK
| | - Aekkachai Tuekprakhon
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Rungtiwa Nutalai
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Beibei Wang
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Guido C Paesen
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, The Wellcome Centre for Human Genetics, Oxford, UK
| | - Cesar Lopez-Camacho
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Jose Slon-Campos
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Bassam Hallis
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, UK
| | - Naomi Coombes
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, UK
| | - Kevin Bewley
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, UK
| | - Sue Charlton
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, UK
| | - Thomas S Walter
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Donal Skelly
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Peter Medawar Building for Pathogen Research, Oxford, UK; Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Sheila F Lumley
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Christina Dold
- NIHR Oxford Biomedical Research Centre, Oxford, UK; Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | | | - Tao Dong
- Chinese Academy of Medical Science (CAMS), Oxford Institute (COI), University of Oxford, Oxford, UK; Nuffield Department of Medicine, University of Oxford, Oxford, UK; MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Andrew J Pollard
- NIHR Oxford Biomedical Research Centre, Oxford, UK; Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Julian C Knight
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS), Oxford Institute (COI), University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Derrick Crook
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Teresa Lambe
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Elizabeth Clutterbuck
- NIHR Oxford Biomedical Research Centre, Oxford, UK; Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Sagida Bibi
- NIHR Oxford Biomedical Research Centre, Oxford, UK; Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Amy Flaxman
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mustapha Bittaye
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Sarah Gilbert
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - William James
- Sir William Dunn School of Pathology University of Oxford, Oxford, UK
| | - Miles W Carroll
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; National Infection Service, Public Health England (PHE), Porton Down, Salisbury, UK
| | - Paul Klenerman
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Peter Medawar Building for Pathogen Research, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK; Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Eleanor Barnes
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Peter Medawar Building for Pathogen Research, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK; Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Susanna J Dunachie
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Peter Medawar Building for Pathogen Research, Oxford, UK; Nuffield Department of Medicine, University of Oxford, Oxford, UK; Centre For Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand
| | - Elizabeth E Fry
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, The Wellcome Centre for Human Genetics, Oxford, UK
| | - Juthathip Mongkolsapaya
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS), Oxford Institute (COI), University of Oxford, Oxford, UK; Siriraj Center of Research Excellence in Dengue & Emerging Pathogens, Dean Office for Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Thailand.
| | - Jingshan Ren
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, The Wellcome Centre for Human Genetics, Oxford, UK.
| | - David I Stuart
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, The Wellcome Centre for Human Genetics, Oxford, UK; Chinese Academy of Medical Science (CAMS), Oxford Institute (COI), University of Oxford, Oxford, UK; Diamond Light Source Ltd, Harwell Science & Innovation Campus, Didcot, UK; Instruct-ERIC, Oxford House, Parkway Court, John Smith Drive, Oxford, UK.
| | - Gavin R Screaton
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS), Oxford Institute (COI), University of Oxford, Oxford, UK.
| |
Collapse
|
680
|
Papasavas P, Olugbile S, Wu U, Robinson K, Roberts AL, O'Sullivan DM, McLaughlin T, Mather JF, Steinberg AC, Orlando R, Kumar A. Seroprevalence of SARS-CoV-2 antibodies, associated epidemiological factors and antibody kinetics among healthcare workers in Connecticut. J Hosp Infect 2021; 114:117-125. [PMID: 33930487 PMCID: PMC8076763 DOI: 10.1016/j.jhin.2021.04.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Healthcare workers (HCWs) are at the front line of the ongoing coronavirus 2019 (COVID-19) pandemic. Comprehensive evaluation of the seroprevalence of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) among HCWs in a large healthcare system could help to identify the impact of epidemiological factors and the presence of symptoms on the immune response to the infection over time. AIM To determine the seroprevalence of SARS-CoV-2-specific antibodies among HCWs, identify associated epidemiological factors and study antibody kinetics. METHODS A longitudinal evaluation of the seroprevalence and epidemiology of SARS-CoV-2-specific antibodies was undertaken in approximately 30,000 HCWs in the largest healthcare system in Connecticut, USA. FINDINGS At baseline, the prevalence of SARS-CoV-2 antibody among 6863 HCWs was 6.3% [95% confidence interval (CI) 5.7-6.9%], and was highest among patient care support (16.7%), medical assistants (9.1%) and nurses (8.2%), and lower for physicians (3.8%) and advanced practice providers (4.5%). Seroprevalence was significantly higher among African Americans [odds ratio (OR) 3.26 compared with Caucasians, 95% CI 1.77-5.99], in participants with at least one symptom of COVID-19 (OR 3.00, 95% CI 1.92-4.68), and in those reporting prior quarantine (OR 3.83, 95% CI 2.57-5.70). No symptoms were reported in 24% of seropositive participants. Among the 47% of participants who returned for a follow-up serological test, the seroreversion rate was 39.5% and the seroconversion rate was 2.2%. The incidence of re-infection in the seropositive group was zero. CONCLUSION Although there is a decline in the immunoglobulin G antibody signal over time, 60.5% of seropositive HCWs had maintained their seroconversion status after a median of 5.5 months.
Collapse
Affiliation(s)
- P Papasavas
- Department of Surgery, Hartford Hospital, Hartford, CT, USA.
| | - S Olugbile
- Cancer Institute, Hartford HealthCare, Hartford, CT, USA
| | - U Wu
- Administration, Hartford HealthCare, Hartford, CT, USA
| | - K Robinson
- Department of Emergency Medicine, Hartford Hospital, Hartford, CT, USA
| | - A L Roberts
- Department of Clinical Laboratory Services: Microbiology, Hartford HealthCare, Hartford, CT, USA
| | | | - T McLaughlin
- Department of Surgery, Hartford Hospital, Hartford, CT, USA
| | - J F Mather
- Hartford Healthcare Research Program, Hartford, CT, USA
| | - A C Steinberg
- Department of Medical Affairs, Hartford HealthCare, Hartford, CT, USA
| | - R Orlando
- Department of Academic Affairs, Hartford HealthCare, Hartford, CT, USA
| | - A Kumar
- Clinical Affairs, Hartford HealthCare, Hartford, CT, USA
| |
Collapse
|
681
|
Choudhry N, Drysdale K, Usai C, Leighton D, Sonagara V, Buchanan R, Nijjar M, Thomas S, Hopkins M, Cutino-Moguel T, Gill US, Foster GR, Kennedy PT. Disparities of SARS-CoV-2 Nucleoprotein-Specific IgG in Healthcare Workers in East London, UK. Front Med (Lausanne) 2021; 8:642723. [PMID: 33987193 PMCID: PMC8111172 DOI: 10.3389/fmed.2021.642723] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/19/2021] [Indexed: 01/17/2023] Open
Abstract
Introduction: SARS-CoV-2 antibody detection serves as an important diagnostic marker for past SARS-CoV-2 infection and is essential to determine the spread of COVID-19, monitor potential COVID-19 long-term effects, and to evaluate possible protection from reinfection. A study was conducted across three hospital sites in a large central London NHS Trust in the UK, to evaluate the prevalence and duration of SARS-CoV-2 IgG antibody positivity in healthcare workers. Methods: A matrix equivalence study consisting of 228 participants was undertaken to evaluate the Abbott Panbio™ COVID-19 IgG/IgM rapid test device. Subsequently, 2001 evaluable healthcare workers (HCW), representing a diverse population, were enrolled in a HCW study between June and August 2020. A plasma sample from each HCW was evaluated using the Abbott Panbio™ COVID-19 IgG/IgM rapid test device, with confirmation of IgG-positive results by the Abbott ArchitectTM SARS-CoV-2 IgG assay. 545 participants, of whom 399 were antibody positive at enrolment, were followed up at 3 months. Results: The Panbio™ COVID-19 IgG/IgM rapid test device demonstrated a high concordance with laboratory tests. SARS-CoV-2 antibodies were detected in 506 participants (25.3%) at enrolment, with a higher prevalence in COVID-19 frontline (28.3%) than non-frontline (19.9%) staff. At follow-up, 274/399 antibody positive participants (68.7%) retained antibodies; 4/146 participants negative at enrolment (2.7%) had seroconverted. Non-white ethnicity, older age, hypertension and COVID-19 symptoms were independent predictors of higher antibody levels (OR 1.881, 2.422-3.034, 2.128, and 1.869 respectively), based on Architect™ index quartiles; participants in the first three categories also showed a greater antibody persistence at 3 months. Conclusion: The SARS-CoV-2 anti-nucleocapsid IgG positivity rate among healthcare staff was high, declining by 31.3% during the 3-month follow-up interval. Interestingly, the IgG-positive participants with certain risk factors for severe COVID-19 illness (older age, Black or Asian Ethnicity hypertension) demonstrated greater persistence over time when compared to the IgG-positive participants without these risk factors.
Collapse
Affiliation(s)
- Naheed Choudhry
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Kate Drysdale
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Barts Health NHS Trust, The Royal London Hospital, London, United Kingdom
| | - Carla Usai
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Dean Leighton
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Vinay Sonagara
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Barts Health NHS Trust, The Royal London Hospital, London, United Kingdom
| | - Ruaridh Buchanan
- Barts Health NHS Trust, Newham General Hospital, London, United Kingdom
| | - Manreet Nijjar
- Barts Health NHS Trust, Whipps Cross Hospital, London, United Kingdom
| | - Sherine Thomas
- Barts Health NHS Trust, Whipps Cross Hospital, London, United Kingdom
| | - Mark Hopkins
- Barts Health NHS Trust, The Royal London Hospital, London, United Kingdom
| | | | - Upkar S. Gill
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Barts Health NHS Trust, The Royal London Hospital, London, United Kingdom
| | - Graham R. Foster
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Barts Health NHS Trust, The Royal London Hospital, London, United Kingdom
| | - Patrick T. Kennedy
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Barts Health NHS Trust, The Royal London Hospital, London, United Kingdom
| |
Collapse
|
682
|
Schiliro M, Vogel ER, Paolini L, Pabelick CM. Cigarette Smoke Exposure, Pediatric Lung Disease, and COVID-19. Front Physiol 2021; 12:652198. [PMID: 33986692 PMCID: PMC8110920 DOI: 10.3389/fphys.2021.652198] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/06/2021] [Indexed: 12/29/2022] Open
Abstract
The detrimental effects of tobacco exposure on children's health are well known. Nonetheless, the prevalence of secondhand or direct cigarette smoke exposure (CSE) in the pediatric population has not significantly decreased over time. On the contrary, the rapid incline in use of e-cigarettes among adolescents has evoked public health concerns since increasing cases of vaping-induced acute lung injury have highlighted the potential harm of these new "smoking" devices. Two pediatric populations are especially vulnerable to the detrimental effects of cigarette smoke. The first group is former premature infants whose risk is elevated both due to their prematurity as well as other risk factors such as oxygen and mechanical ventilation to which they are disproportionately exposed. The second group is children and adolescents with chronic respiratory diseases, in particular asthma and other wheezing disorders. Coronavirus disease 2019 (COVID-19) is a spectrum of diseases caused by infection with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that has spread worldwide over the last year. Here, respiratory symptoms ranging from mild to acute respiratory distress syndrome (ARDS) are at the forefront of COVID-19 cases among adults, and cigarette smoking is associated with worse outcomes in this population, and cigarette smoking is associated with worse outcomes in this population. Interestingly, SARS-CoV-2 infection affects children differently in regard to infection susceptibility, disease manifestations, and complications. Although children carry and transmit the virus, the likelihood of symptomatic infection is low, and the rates of hospitalization and death are even lower when compared to the adult population. However, multisystem inflammatory syndrome is recognized as a serious consequence of SARS-CoV-2 infection in the pediatric population. In addition, recent data demonstrate specific clinical patterns in children infected with SARS-CoV-2 who develop multisystem inflammatory syndrome vs. severe COVID-19. In this review, we highlight the pulmonary effects of CSE in vulnerable pediatric populations in the context of the ongoing SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Marta Schiliro
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Elizabeth R. Vogel
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Lucia Paolini
- Department of Pediatric, San Gerardo Hospital, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Christina M. Pabelick
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
683
|
Dispinseri S, Lampasona V, Secchi M, Cara A, Bazzigaluppi E, Negri D, Brigatti C, Pirillo MF, Marzinotto I, Borghi M, Rovere-Querini P, Tresoldi C, Ciceri F, Scavini M, Scarlatti G, Piemonti L. Robust Neutralizing Antibodies to SARS-CoV-2 Develop and Persist in Subjects with Diabetes and COVID-19 Pneumonia. J Clin Endocrinol Metab 2021; 106:1472-1481. [PMID: 33513242 PMCID: PMC7928901 DOI: 10.1210/clinem/dgab055] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Indexed: 12/21/2022]
Abstract
CONTEXT Demonstrating the ability to mount a neutralizing antibody response to SARS-CoV-2 in the presence of diabetes is crucial to understand COVID-19 pathogenesis, reinfection potential, and vaccine development. OBJECTIVE The aim of this study was to characterize the kinetics and durability of neutralizing antibody (Nab) response against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the presence of hyperglycemia. METHODS Using a lentiviral vector-based SARS-CoV-2 neutralization assay to measure Nabs, we characterized 150 patients randomly selected from a cohort of 509 patients with confirmed COVID-19 pneumonia. We analyzed Nab response according to the presence of diabetes or hyperglycemia, at the time of hospitalization and during the postdischarge follow-up: 1-, 3-, and 6-month outpatient visits. RESULTS Among 150 randomly selected patients 40 (26.6%) had diabetes. Diabetes (hazard ratio [HR] 8.9, P < .001), glucose levels (HR 1.25 × 1.1 mmol/L, P < .001), and glucose variability (HR 1.17 × 0.6 mmol/L, P < .001) were independently associated with an increased risk of mortality. The neutralizing activity of SARS-CoV-2 antibodies in patients with diabetes was superimposable, as for kinetics and extent, to that of patients without diabetes. It was similar across glucose levels and correlated with the humoral response against the SARS-CoV-2 spike protein. Positivity for Nabs at the time of hospital admission conferred protection on mortality, both in the presence (HR 0.28, P = .046) or absence of diabetes (HR 0.26, P = .030). The longevity of the Nab response was not affected by diabetes. CONCLUSION Diabetes and hyperglycemia do not affect the kinetics and durability of the neutralizing antibody response to SARS-CoV-2. These findings provide the rational to include patients with diabetes in the early phase of the vaccination campaign against SARS-CoV-2.
Collapse
Affiliation(s)
- Stefania Dispinseri
- Viral Evolution and Trasmission Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Vito Lampasona
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Andrea Cara
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | | | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Cristina Brigatti
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Ilaria Marzinotto
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Martina Borghi
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Patrizia Rovere-Querini
- Department of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
- School of Medicine and Surgery, Università Vita-Salute San Raffaele, Milan, Italy
| | - Cristina Tresoldi
- Molecular Hematology Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Fabio Ciceri
- School of Medicine and Surgery, Università Vita-Salute San Raffaele, Milan, Italy
- Hematology and Bone Marrow Transplantation Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Marina Scavini
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Gabriella Scarlatti
- Viral Evolution and Trasmission Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- School of Medicine and Surgery, Università Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
684
|
Bortz RH, Florez C, Laudermilch E, Wirchnianski AS, Lasso G, Malonis RJ, Georgiev GI, Vergnolle O, Herrera NG, Morano NC, Campbell ST, Orner EP, Mengotto A, Dieterle ME, Fels JM, Haslwanter D, Jangra RK, Celikgil A, Kimmel D, Lee JH, Mariano MC, Nakouzi A, Quiroz J, Rivera J, Szymczak WA, Tong K, Barnhill J, Forsell MNE, Ahlm C, Stein DT, Pirofski LA, Goldstein DY, Garforth SJ, Almo SC, Daily JP, Prystowsky MB, Faix JD, Fox AS, Weiss LM, Lai JR, Chandran K. Single-Dilution COVID-19 Antibody Test with Qualitative and Quantitative Readouts. mSphere 2021; 6:e00224-21. [PMID: 33883259 PMCID: PMC8546701 DOI: 10.1128/msphere.00224-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 03/24/2021] [Indexed: 12/24/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) global pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to place an immense burden on societies and health care systems. A key component of COVID-19 control efforts is serological testing to determine the community prevalence of SARS-CoV-2 exposure and quantify individual immune responses to prior SARS-CoV-2 infection or vaccination. Here, we describe a laboratory-developed antibody test that uses readily available research-grade reagents to detect SARS-CoV-2 exposure in patient blood samples with high sensitivity and specificity. We further show that this sensitive test affords the estimation of viral spike-specific IgG titers from a single sample measurement, thereby providing a simple and scalable method to measure the strength of an individual's immune response. The accuracy, adaptability, and cost-effectiveness of this test make it an excellent option for clinical deployment in the ongoing COVID-19 pandemic.IMPORTANCE Serological surveillance has become an important public health tool during the COVID-19 pandemic. Detection of protective antibodies and seroconversion after SARS-CoV-2 infection or vaccination can help guide patient care plans and public health policies. Serology tests can detect antibodies against past infections; consequently, they can help overcome the shortcomings of molecular tests, which can detect only active infections. This is important, especially when considering that many COVID-19 patients are asymptomatic. In this study, we describe an enzyme-linked immunosorbent assay (ELISA)-based qualitative and quantitative serology test developed to measure IgG and IgA antibodies against the SARS-CoV-2 spike glycoprotein. The test can be deployed using commonly available laboratory reagents and equipment and displays high specificity and sensitivity. Furthermore, we demonstrate that IgG titers in patient samples can be estimated from a single measurement, enabling the assay's use in high-throughput clinical environments.
Collapse
Affiliation(s)
- Robert H Bortz
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Catalina Florez
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Chemistry and Life Science, United States Military Academy at West Point, West Point, New York, USA
| | - Ethan Laudermilch
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Ariel S Wirchnianski
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Gorka Lasso
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Ryan J Malonis
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - George I Georgiev
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Olivia Vergnolle
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Natalia G Herrera
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Nicholas C Morano
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Sean T Campbell
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
- Montefiore Medical Center, Bronx, New York, USA
| | - Erika P Orner
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
- Montefiore Medical Center, Bronx, New York, USA
| | - Amanda Mengotto
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
- Montefiore Medical Center, Bronx, New York, USA
| | - M Eugenia Dieterle
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - J Maximilian Fels
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Denise Haslwanter
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Rohit K Jangra
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Alev Celikgil
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Duncan Kimmel
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
- Montefiore Medical Center, Bronx, New York, USA
| | - James H Lee
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Margarette C Mariano
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Antonio Nakouzi
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
- Montefiore Medical Center, Bronx, New York, USA
| | - Jose Quiroz
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
- Montefiore Medical Center, Bronx, New York, USA
| | - Johanna Rivera
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
- Montefiore Medical Center, Bronx, New York, USA
| | - Wendy A Szymczak
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
- Montefiore Medical Center, Bronx, New York, USA
| | - Karen Tong
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jason Barnhill
- Department of Chemistry and Life Science, United States Military Academy at West Point, West Point, New York, USA
| | | | - Clas Ahlm
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Daniel T Stein
- Montefiore Medical Center, Bronx, New York, USA
- Division of Endocrinology and Diabetes, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Liise-Anne Pirofski
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
- Montefiore Medical Center, Bronx, New York, USA
| | - D Yitzchak Goldstein
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
- Montefiore Medical Center, Bronx, New York, USA
| | - Scott J Garforth
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Steven C Almo
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Johanna P Daily
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
- Montefiore Medical Center, Bronx, New York, USA
| | - Michael B Prystowsky
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
- Montefiore Medical Center, Bronx, New York, USA
| | - James D Faix
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
- Montefiore Medical Center, Bronx, New York, USA
| | - Amy S Fox
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
- Montefiore Medical Center, Bronx, New York, USA
| | - Louis M Weiss
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
- Montefiore Medical Center, Bronx, New York, USA
| | - Jonathan R Lai
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
685
|
Hoffmann M, Zhang L, Krüger N, Graichen L, Kleine-Weber H, Hofmann-Winkler H, Kempf A, Nessler S, Riggert J, Winkler MS, Schulz S, Jäck HM, Pöhlmann S. SARS-CoV-2 mutations acquired in mink reduce antibody-mediated neutralization. Cell Rep 2021; 35:109017. [PMID: 33857422 PMCID: PMC8018833 DOI: 10.1016/j.celrep.2021.109017] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/16/2021] [Accepted: 03/30/2021] [Indexed: 01/01/2023] Open
Abstract
Transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) from humans to farmed mink has been observed in Europe and the US. In the infected animals, viral variants arose that harbored mutations in the spike (S) protein, the target of neutralizing antibodies, and these variants were transmitted back to humans. This raised concerns that mink might become a constant source of human infection with SARS-CoV-2 variants associated with an increased threat to human health and resulted in mass culling of mink. Here, we report that mutations frequently found in the S proteins of SARS-CoV-2 from mink are mostly compatible with efficient entry into human cells and its inhibition by soluble angiotensin-converting enzyme 2 (ACE2). In contrast, mutation Y453F reduces neutralization by an antibody with emergency use authorization for coronavirus disease 2019 (COVID-19) therapy and sera/plasma from COVID-19 patients. These results suggest that antibody responses induced upon infection or certain antibodies used for treatment might offer insufficient protection against SARS-CoV-2 variants from mink.
Collapse
Affiliation(s)
- Markus Hoffmann
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany; Faculty of Biology and Psychology, University Göttingen, Wilhelmsplatz 1, 37073 Göttingen, Germany.
| | - Lu Zhang
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany; Faculty of Biology and Psychology, University Göttingen, Wilhelmsplatz 1, 37073 Göttingen, Germany
| | - Nadine Krüger
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Luise Graichen
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Hannah Kleine-Weber
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany; Faculty of Biology and Psychology, University Göttingen, Wilhelmsplatz 1, 37073 Göttingen, Germany
| | - Heike Hofmann-Winkler
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Amy Kempf
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany; Faculty of Biology and Psychology, University Göttingen, Wilhelmsplatz 1, 37073 Göttingen, Germany
| | - Stefan Nessler
- Institute of Neuropathology, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Joachim Riggert
- Department of Transfusion Medicine, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Martin Sebastian Winkler
- Department of Anaesthesiology and Intensive Care Medicine, University of Göttingen Medical Center, Göttingen, Georg-August University of Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Sebastian Schulz
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander University of Erlangen-Nürnberg, Glückstraße 6, 91054 Erlangen, Germany
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander University of Erlangen-Nürnberg, Glückstraße 6, 91054 Erlangen, Germany
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany; Faculty of Biology and Psychology, University Göttingen, Wilhelmsplatz 1, 37073 Göttingen, Germany.
| |
Collapse
|
686
|
Garrido C, Hurst JH, Lorang CG, Aquino JN, Rodriguez J, Pfeiffer TS, Singh T, Semmes EC, Lugo DJ, Rotta AT, Turner NA, Burke TW, McClain MT, Petzold EA, Permar SR, Moody MA, Woods CW, Kelly MS, Fouda GG. Asymptomatic or mild symptomatic SARS-CoV-2 infection elicits durable neutralizing antibody responses in children and adolescents. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021. [PMID: 33907760 DOI: 10.1101/2021.04.17.21255663] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
As SARS-CoV-2 continues to spread globally, questions have emerged regarding the strength and durability of immune responses in specific populations. In this study, we evaluated humoral immune responses in 69 children and adolescents with asymptomatic or mild symptomatic SARS-CoV-2 infection. We detected robust IgM, IgG, and IgA antibody responses to a broad array of SARS-CoV-2 antigens at the time of acute infection and 2 and 4 months after acute infection in all participants. Notably, these antibody responses were associated with virus neutralizing activity that was still detectable 4 months after acute infection in 94% of children. Moreover, antibody responses and neutralizing activity in sera from children and adolescents were comparable or superior to those observed in sera from 24 adults with mild symptomatic infection. Taken together, these findings indicate children and adolescents with mild or asymptomatic SARS-CoV-2 infection generate robust and durable humoral immune responses that are likely to protect from reinfection.
Collapse
|
687
|
Jangra S, De Vrieze J, Choi A, Rathnasinghe R, Laghlali G, Uvyn A, Van Herck S, Nuhn L, Deswarte K, Zhong Z, Sanders NN, Lienenklaus S, David SA, Strohmeier S, Amanat F, Krammer F, Hammad H, Lambrecht BN, Coughlan L, García‐Sastre A, De Geest BG, Schotsaert M. Sterilizing Immunity against SARS-CoV-2 Infection in Mice by a Single-Shot and Lipid Amphiphile Imidazoquinoline TLR7/8 Agonist-Adjuvanted Recombinant Spike Protein Vaccine*. Angew Chem Int Ed Engl 2021; 60:9467-9473. [PMID: 33464672 PMCID: PMC8014308 DOI: 10.1002/anie.202015362] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/24/2020] [Indexed: 12/21/2022]
Abstract
The search for vaccines that protect from severe morbidity and mortality because of infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes coronavirus disease 2019 (COVID-19) is a race against the clock and the virus. Here we describe an amphiphilic imidazoquinoline (IMDQ-PEG-CHOL) TLR7/8 adjuvant, consisting of an imidazoquinoline conjugated to the chain end of a cholesterol-poly(ethylene glycol) macromolecular amphiphile. It is water-soluble and exhibits massive translocation to lymph nodes upon local administration through binding to albumin, affording localized innate immune activation and reduction in systemic inflammation. The adjuvanticity of IMDQ-PEG-CHOL was validated in a licensed vaccine setting (quadrivalent influenza vaccine) and an experimental trimeric recombinant SARS-CoV-2 spike protein vaccine, showing robust IgG2a and IgG1 antibody titers in mice that could neutralize viral infection in vitro and in vivo in a mouse model.
Collapse
Affiliation(s)
- Sonia Jangra
- Department of MicrobiologyIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | | | - Angela Choi
- Department of MicrobiologyIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Graduate School of Biomedical SciencesIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Raveen Rathnasinghe
- Department of MicrobiologyIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Graduate School of Biomedical SciencesIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Gabriel Laghlali
- Department of MicrobiologyIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Annemiek Uvyn
- Department of PharmaceuticsGhent UniversityGhentBelgium
| | | | - Lutz Nuhn
- Max Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| | - Kim Deswarte
- Department of Internal Medicine and PediatricsGhent University and VIB Center for Inflammation ResearchZwijnaardeBelgium
| | - Zifu Zhong
- Department of PharmaceuticsGhent UniversityGhentBelgium
| | | | - Stefan Lienenklaus
- Institute for Laboratory Animal ScienceInstitute of ImmunologyHannover Medical SchoolHannoverGermany
| | | | - Shirin Strohmeier
- Department of MicrobiologyIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Graduate School of Biomedical SciencesIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Fatima Amanat
- Department of MicrobiologyIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Graduate School of Biomedical SciencesIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Florian Krammer
- Department of MicrobiologyIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Hamida Hammad
- Department of Internal Medicine and PediatricsGhent University and VIB Center for Inflammation ResearchZwijnaardeBelgium
| | - Bart N. Lambrecht
- Department of Internal Medicine and PediatricsGhent University and VIB Center for Inflammation ResearchZwijnaardeBelgium
- Department of Pulmonary MedicineErasmus Medical CenterRotterdamThe Netherlands
| | - Lynda Coughlan
- Department of MicrobiologyIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Adolfo García‐Sastre
- Department of MicrobiologyIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Global Health and Emerging Pathogen InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Department of MedicineDivision of Infectious DiseasesIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- The Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | | | - Michael Schotsaert
- Department of MicrobiologyIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Global Health and Emerging Pathogen InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| |
Collapse
|
688
|
Gouma S, Weirick ME, Bolton MJ, Arevalo CP, Goodwin EC, Anderson EM, McAllister CM, Christensen SR, Dunbar D, Fiore D, Brock A, Weaver J, Millar J, DerOhannessian S, Frank I, Rader DJ, Wherry EJ, Hensley SE. Sero-monitoring of health care workers reveals complex relationships between common coronavirus antibodies and SARS-CoV-2 severity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.04.12.21255324. [PMID: 33907765 PMCID: PMC8077588 DOI: 10.1101/2021.04.12.21255324] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Recent common coronavirus (CCV) infections are associated with reduced COVID-19 severity upon SARS-CoV-2 infection, however the immunological mechanisms involved are unknown. We completed serological assays using samples collected from health care workers to identify antibody types associated with SARS-CoV-2 protection and COVID-19 severity. Rare SARS-CoV-2 cross-reactive antibodies elicited by past CCV infections were not associated with protection; however, the duration of symptoms following SARS-CoV-2 infections was significantly reduced in individuals with higher common betacoronavirus (βCoV) antibody titers. Since antibody titers decline over time after CCV infections, individuals in our cohort with higher βCoV antibody titers were more likely recently infected with common βCoVs compared to individuals with lower antibody titers. Therefore, our data suggest that recent βCoV infections potentially limit the severity of SARS-CoV-2 infections through mechanisms that do not involve cross-reactive antibodies. Our data are consistent with the emerging hypothesis that cellular immune responses elicited by recent common βCoV infections transiently reduce disease severity following SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Sigrid Gouma
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Madison E. Weirick
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Marcus J. Bolton
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Claudia P. Arevalo
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Eileen C. Goodwin
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Elizabeth M. Anderson
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Christopher M. McAllister
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Shannon R. Christensen
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Debora Dunbar
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Danielle Fiore
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Amanda Brock
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - JoEllen Weaver
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - John Millar
- Departments of Genetics and Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Stephanie DerOhannessian
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Departments of Genetics and Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - The UPenn COVID Processing Unit
- The UPenn COVID Processing Unit is a unit of individuals from diverse laboratories at the University of Pennsylvania who volunteered time and effort to enable study of COVID-19 patients during the pandemic. Members are listed in the acknowledgement section
| | - Ian Frank
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Daniel J. Rader
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Departments of Genetics and Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - E. John Wherry
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Scott E. Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
689
|
COVID-19 dynamics after a national immunization program in Israel. Nat Med 2021; 27:1055-1061. [PMID: 33875890 DOI: 10.1038/s41591-021-01337-2] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/05/2021] [Indexed: 01/19/2023]
Abstract
Studies on the real-life effect of the BNT162b2 vaccine for Coronavirus Disease 2019 (COVID-19) prevention are urgently needed. In this study, we conducted a retrospective analysis of data from the Israeli Ministry of Health collected between 28 August 2020 and 24 February 2021. We studied the temporal dynamics of the number of new COVID-19 cases and hospitalizations after the vaccination campaign, which was initiated on 20 December 2020. To distinguish the possible effects of the vaccination on cases and hospitalizations from other factors, including a third lockdown implemented on 8 January 2021, we performed several comparisons: (1) individuals aged 60 years and older prioritized to receive the vaccine first versus younger age groups; (2) the January lockdown versus the September lockdown; and (3) early-vaccinated versus late-vaccinated cities. A larger and earlier decrease in COVID-19 cases and hospitalization was observed in individuals older than 60 years, followed by younger age groups, by the order of vaccination prioritization. This pattern was not observed in the previous lockdown and was more pronounced in early-vaccinated cities. Our analysis demonstrates the real-life effect of a national vaccination campaign on the pandemic dynamics.
Collapse
|
690
|
Hall VJ, Foulkes S, Charlett A, Atti A, Monk EJM, Simmons R, Wellington E, Cole MJ, Saei A, Oguti B, Munro K, Wallace S, Kirwan PD, Shrotri M, Vusirikala A, Rokadiya S, Kall M, Zambon M, Ramsay M, Brooks T, Brown CS, Chand MA, Hopkins S. SARS-CoV-2 infection rates of antibody-positive compared with antibody-negative health-care workers in England: a large, multicentre, prospective cohort study (SIREN). Lancet 2021; 397:1459-1469. [PMID: 33844963 DOI: 10.1101/2021.01.13.21249642] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/01/2021] [Accepted: 03/12/2021] [Indexed: 05/27/2023]
Abstract
BACKGROUND Increased understanding of whether individuals who have recovered from COVID-19 are protected from future SARS-CoV-2 infection is an urgent requirement. We aimed to investigate whether antibodies against SARS-CoV-2 were associated with a decreased risk of symptomatic and asymptomatic reinfection. METHODS A large, multicentre, prospective cohort study was done, with participants recruited from publicly funded hospitals in all regions of England. All health-care workers, support staff, and administrative staff working at hospitals who could remain engaged in follow-up for 12 months were eligible to join The SARS-CoV-2 Immunity and Reinfection Evaluation study. Participants were excluded if they had no PCR tests after enrolment, enrolled after Dec 31, 2020, or had insufficient PCR and antibody data for cohort assignment. Participants attended regular SARS-CoV-2 PCR and antibody testing (every 2-4 weeks) and completed questionnaires every 2 weeks on symptoms and exposures. At enrolment, participants were assigned to either the positive cohort (antibody positive, or previous positive PCR or antibody test) or negative cohort (antibody negative, no previous positive PCR or antibody test). The primary outcome was a reinfection in the positive cohort or a primary infection in the negative cohort, determined by PCR tests. Potential reinfections were clinically reviewed and classified according to case definitions (confirmed, probable, or possible) and symptom-status, depending on the hierarchy of evidence. Primary infections in the negative cohort were defined as a first positive PCR test and seroconversions were excluded when not associated with a positive PCR test. A proportional hazards frailty model using a Poisson distribution was used to estimate incidence rate ratios (IRR) to compare infection rates in the two cohorts. FINDINGS From June 18, 2020, to Dec 31, 2020, 30 625 participants were enrolled into the study. 51 participants withdrew from the study, 4913 were excluded, and 25 661 participants (with linked data on antibody and PCR testing) were included in the analysis. Data were extracted from all sources on Feb 5, 2021, and include data up to and including Jan 11, 2021. 155 infections were detected in the baseline positive cohort of 8278 participants, collectively contributing 2 047 113 person-days of follow-up. This compares with 1704 new PCR positive infections in the negative cohort of 17 383 participants, contributing 2 971 436 person-days of follow-up. The incidence density was 7·6 reinfections per 100 000 person-days in the positive cohort, compared with 57·3 primary infections per 100 000 person-days in the negative cohort, between June, 2020, and January, 2021. The adjusted IRR was 0·159 for all reinfections (95% CI 0·13-0·19) compared with PCR-confirmed primary infections. The median interval between primary infection and reinfection was more than 200 days. INTERPRETATION A previous history of SARS-CoV-2 infection was associated with an 84% lower risk of infection, with median protective effect observed 7 months following primary infection. This time period is the minimum probable effect because seroconversions were not included. This study shows that previous infection with SARS-CoV-2 induces effective immunity to future infections in most individuals. FUNDING Department of Health and Social Care of the UK Government, Public Health England, The National Institute for Health Research, with contributions from the Scottish, Welsh and Northern Irish governments.
Collapse
Affiliation(s)
- Victoria Jane Hall
- Public Health England Colindale, Colindale, London, UK; The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, University of Oxford, Oxford, UK
| | - Sarah Foulkes
- Public Health England Colindale, Colindale, London, UK
| | - Andre Charlett
- Public Health England Colindale, Colindale, London, UK; The National Institute for Health Research Health Protection Research Unit in Behavioural Science and Evaluation at University of Bristol in partnership with Public Health England, Bristol, UK
| | - Ana Atti
- Public Health England Colindale, Colindale, London, UK
| | | | - Ruth Simmons
- Public Health England Colindale, Colindale, London, UK
| | | | | | - Ayoub Saei
- Public Health England Colindale, Colindale, London, UK
| | - Blanche Oguti
- Public Health England Colindale, Colindale, London, UK; Oxford Vaccine Group, University of Oxford, Oxford, UK
| | - Katie Munro
- Public Health England Colindale, Colindale, London, UK
| | - Sarah Wallace
- Public Health England Colindale, Colindale, London, UK
| | - Peter D Kirwan
- Public Health England Colindale, Colindale, London, UK; Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK
| | | | | | | | - Meaghan Kall
- Public Health England Colindale, Colindale, London, UK
| | - Maria Zambon
- Public Health England Colindale, Colindale, London, UK
| | - Mary Ramsay
- Public Health England Colindale, Colindale, London, UK; Oxford Vaccine Group, University of Oxford, Oxford, UK
| | - Tim Brooks
- Public Health England Colindale, Colindale, London, UK
| | - Colin S Brown
- Public Health England Colindale, Colindale, London, UK
| | - Meera A Chand
- Public Health England Colindale, Colindale, London, UK; Guys and St Thomas's Hospital NHS Trust, London, UK
| | - Susan Hopkins
- Public Health England Colindale, Colindale, London, UK; The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, University of Oxford, Oxford, UK.
| |
Collapse
|
691
|
Hall VJ, Foulkes S, Charlett A, Atti A, Monk EJM, Simmons R, Wellington E, Cole MJ, Saei A, Oguti B, Munro K, Wallace S, Kirwan PD, Shrotri M, Vusirikala A, Rokadiya S, Kall M, Zambon M, Ramsay M, Brooks T, Brown CS, Chand MA, Hopkins S. SARS-CoV-2 infection rates of antibody-positive compared with antibody-negative health-care workers in England: a large, multicentre, prospective cohort study (SIREN). Lancet 2021; 397:1459-1469. [PMID: 33844963 PMCID: PMC8040523 DOI: 10.1016/s0140-6736(21)00675-9] [Citation(s) in RCA: 424] [Impact Index Per Article: 106.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/01/2021] [Accepted: 03/12/2021] [Indexed: 01/19/2023]
Abstract
BACKGROUND Increased understanding of whether individuals who have recovered from COVID-19 are protected from future SARS-CoV-2 infection is an urgent requirement. We aimed to investigate whether antibodies against SARS-CoV-2 were associated with a decreased risk of symptomatic and asymptomatic reinfection. METHODS A large, multicentre, prospective cohort study was done, with participants recruited from publicly funded hospitals in all regions of England. All health-care workers, support staff, and administrative staff working at hospitals who could remain engaged in follow-up for 12 months were eligible to join The SARS-CoV-2 Immunity and Reinfection Evaluation study. Participants were excluded if they had no PCR tests after enrolment, enrolled after Dec 31, 2020, or had insufficient PCR and antibody data for cohort assignment. Participants attended regular SARS-CoV-2 PCR and antibody testing (every 2-4 weeks) and completed questionnaires every 2 weeks on symptoms and exposures. At enrolment, participants were assigned to either the positive cohort (antibody positive, or previous positive PCR or antibody test) or negative cohort (antibody negative, no previous positive PCR or antibody test). The primary outcome was a reinfection in the positive cohort or a primary infection in the negative cohort, determined by PCR tests. Potential reinfections were clinically reviewed and classified according to case definitions (confirmed, probable, or possible) and symptom-status, depending on the hierarchy of evidence. Primary infections in the negative cohort were defined as a first positive PCR test and seroconversions were excluded when not associated with a positive PCR test. A proportional hazards frailty model using a Poisson distribution was used to estimate incidence rate ratios (IRR) to compare infection rates in the two cohorts. FINDINGS From June 18, 2020, to Dec 31, 2020, 30 625 participants were enrolled into the study. 51 participants withdrew from the study, 4913 were excluded, and 25 661 participants (with linked data on antibody and PCR testing) were included in the analysis. Data were extracted from all sources on Feb 5, 2021, and include data up to and including Jan 11, 2021. 155 infections were detected in the baseline positive cohort of 8278 participants, collectively contributing 2 047 113 person-days of follow-up. This compares with 1704 new PCR positive infections in the negative cohort of 17 383 participants, contributing 2 971 436 person-days of follow-up. The incidence density was 7·6 reinfections per 100 000 person-days in the positive cohort, compared with 57·3 primary infections per 100 000 person-days in the negative cohort, between June, 2020, and January, 2021. The adjusted IRR was 0·159 for all reinfections (95% CI 0·13-0·19) compared with PCR-confirmed primary infections. The median interval between primary infection and reinfection was more than 200 days. INTERPRETATION A previous history of SARS-CoV-2 infection was associated with an 84% lower risk of infection, with median protective effect observed 7 months following primary infection. This time period is the minimum probable effect because seroconversions were not included. This study shows that previous infection with SARS-CoV-2 induces effective immunity to future infections in most individuals. FUNDING Department of Health and Social Care of the UK Government, Public Health England, The National Institute for Health Research, with contributions from the Scottish, Welsh and Northern Irish governments.
Collapse
Affiliation(s)
- Victoria Jane Hall
- Public Health England Colindale, Colindale, London, UK; The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, University of Oxford, Oxford, UK
| | - Sarah Foulkes
- Public Health England Colindale, Colindale, London, UK
| | - Andre Charlett
- Public Health England Colindale, Colindale, London, UK; The National Institute for Health Research Health Protection Research Unit in Behavioural Science and Evaluation at University of Bristol in partnership with Public Health England, Bristol, UK
| | - Ana Atti
- Public Health England Colindale, Colindale, London, UK
| | | | - Ruth Simmons
- Public Health England Colindale, Colindale, London, UK
| | | | | | - Ayoub Saei
- Public Health England Colindale, Colindale, London, UK
| | - Blanche Oguti
- Public Health England Colindale, Colindale, London, UK; Oxford Vaccine Group, University of Oxford, Oxford, UK
| | - Katie Munro
- Public Health England Colindale, Colindale, London, UK
| | - Sarah Wallace
- Public Health England Colindale, Colindale, London, UK
| | - Peter D Kirwan
- Public Health England Colindale, Colindale, London, UK; Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK
| | | | | | | | - Meaghan Kall
- Public Health England Colindale, Colindale, London, UK
| | - Maria Zambon
- Public Health England Colindale, Colindale, London, UK
| | - Mary Ramsay
- Public Health England Colindale, Colindale, London, UK; Oxford Vaccine Group, University of Oxford, Oxford, UK
| | - Tim Brooks
- Public Health England Colindale, Colindale, London, UK
| | - Colin S Brown
- Public Health England Colindale, Colindale, London, UK
| | - Meera A Chand
- Public Health England Colindale, Colindale, London, UK; Guys and St Thomas's Hospital NHS Trust, London, UK
| | - Susan Hopkins
- Public Health England Colindale, Colindale, London, UK; The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, University of Oxford, Oxford, UK.
| |
Collapse
|
692
|
Chvatal-Medina M, Mendez-Cortina Y, Patiño PJ, Velilla PA, Rugeles MT. Antibody Responses in COVID-19: A Review. Front Immunol 2021; 12:633184. [PMID: 33936045 PMCID: PMC8081880 DOI: 10.3389/fimmu.2021.633184] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/25/2021] [Indexed: 01/08/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to spread worldwide as a severe pandemic. Although its seroprevalence is highly variable among territories, it has been reported at around 10%, but higher in health workers. Evidence regarding cross-neutralizing response between SARS-CoV and SARS-CoV-2 is still controversial. However, other previous coronaviruses may interfere with SARS-CoV-2 infection, since they are phylogenetically related and share the same target receptor. Further, the seroconversion of IgM and IgG occurs at around 12 days post onset of symptoms and most patients have neutralizing titers on days 14-20, with great titer variability. Neutralizing antibodies correlate positively with age, male sex, and severity of the disease. Moreover, the use of convalescent plasma has shown controversial results in terms of safety and efficacy, and due to the variable immune response among individuals, measuring antibody titers before transfusion is mostly required. Similarly, cellular immunity seems to be crucial in the resolution of the infection, as SARS-CoV-2-specific CD4+ and CD8+ T cells circulate to some extent in recovered patients. Of note, the duration of the antibody response has not been well established yet.
Collapse
Affiliation(s)
- Mateo Chvatal-Medina
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | | | - Pablo J. Patiño
- Grupo Inmunodeficiencias Primarias, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Paula A. Velilla
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Maria T. Rugeles
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
693
|
Letizia AG, Ge Y, Vangeti S, Goforth C, Weir DL, Kuzmina NA, Balinsky CA, Chen HW, Ewing D, Soares-Schanoski A, George MC, Graham WD, Jones F, Bharaj P, Lizewski RA, Lizewski SE, Marayag J, Marjanovic N, Miller CM, Mofsowitz S, Nair VD, Nunez E, Parent DM, Porter CK, Santa Ana E, Schilling M, Stadlbauer D, Sugiharto VA, Termini M, Sun P, Tracy RP, Krammer F, Bukreyev A, Ramos I, Sealfon SC. SARS-CoV-2 seropositivity and subsequent infection risk in healthy young adults: a prospective cohort study. THE LANCET RESPIRATORY MEDICINE 2021; 9:712-720. [PMID: 33865504 PMCID: PMC8049591 DOI: 10.1016/s2213-2600(21)00158-2] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/09/2021] [Accepted: 03/15/2021] [Indexed: 01/23/2023]
Abstract
Background Whether young adults who are infected with SARS-CoV-2 are at risk of subsequent infection is uncertain. We investigated the risk of subsequent SARS-CoV-2 infection among young adults seropositive for a previous infection. Methods This analysis was performed as part of the prospective COVID-19 Health Action Response for Marines study (CHARM). CHARM included predominantly male US Marine recruits, aged 18–20 years, following a 2-week unsupervised quarantine at home. After the home quarantine period, upon arrival at a Marine-supervised 2-week quarantine facility (college campus or hotel), participants were enrolled and were assessed for baseline SARS-CoV-2 IgG seropositivity, defined as a dilution of 1:150 or more on receptor-binding domain and full-length spike protein ELISA. Participants also completed a questionnaire consisting of demographic information, risk factors, reporting of 14 specific COVID-19-related symptoms or any other unspecified symptom, and brief medical history. SARS-CoV-2 infection was assessed by PCR at weeks 0, 1, and 2 of quarantine and participants completed a follow-up questionnaire, which included questions about the same COVID-19-related symptoms since the last study visit. Participants were excluded at this stage if they had a positive PCR test during quarantine. Participants who had three negative swab PCR results during quarantine and a baseline serum serology test at the beginning of the supervised quarantine that identified them as seronegative or seropositive for SARS-CoV-2 then went on to basic training at Marine Corps Recruit Depot—Parris Island. Three PCR tests were done at weeks 2, 4, and 6 in both seropositive and seronegative groups, along with the follow-up symptom questionnaire and baseline neutralising antibody titres on all subsequently infected seropositive and selected seropositive uninfected participants (prospective study period). Findings Between May 11, 2020, and Nov 2, 2020, we enrolled 3249 participants, of whom 3168 (98%) continued into the 2-week quarantine period. 3076 (95%) participants, 2825 (92%) of whom were men, were then followed up during the prospective study period after quarantine for 6 weeks. Among 189 seropositive participants, 19 (10%) had at least one positive PCR test for SARS-CoV-2 during the 6-week follow-up (1·1 cases per person-year). In contrast, 1079 (48%) of 2247 seronegative participants tested positive (6·2 cases per person-year). The incidence rate ratio was 0·18 (95% CI 0·11–0·28; p<0·001). Among seropositive recruits, infection was more likely with lower baseline full-length spike protein IgG titres than in those with higher baseline full-length spike protein IgG titres (hazard ratio 0·45 [95% CI 0·32–0·65]; p<0·001). Infected seropositive participants had viral loads that were about 10-times lower than those of infected seronegative participants (ORF1ab gene cycle threshold difference 3·95 [95% CI 1·23–6·67]; p=0·004). Among seropositive participants, baseline neutralising titres were detected in 45 (83%) of 54 uninfected and in six (32%) of 19 infected participants during the 6 weeks of observation (ID50 difference p<0·0001). Interpretation Seropositive young adults had about one-fifth the risk of subsequent infection compared with seronegative individuals. Although antibodies induced by initial infection are largely protective, they do not guarantee effective SARS-CoV-2 neutralisation activity or immunity against subsequent infection. These findings might be relevant for optimisation of mass vaccination strategies. Funding Defense Health Agency and Defense Advanced Research Projects Agency.
Collapse
Affiliation(s)
| | - Yongchao Ge
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sindhu Vangeti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carl Goforth
- Naval Medical Research Center, Silver Spring, MD, USA
| | - Dawn L Weir
- Naval Medical Research Center, Silver Spring, MD, USA
| | - Natalia A Kuzmina
- Department of Pathology University of Texas Medical Branch and Galveston National Laboratory, Galveston, TX, USA
| | | | - Hua Wei Chen
- Naval Medical Research Center, Silver Spring, MD, USA
| | - Dan Ewing
- Naval Medical Research Center, Silver Spring, MD, USA
| | | | | | | | - Franca Jones
- Naval Medical Research Center, Silver Spring, MD, USA
| | - Preeti Bharaj
- Department of Pathology University of Texas Medical Branch and Galveston National Laboratory, Galveston, TX, USA
| | | | | | - Jan Marayag
- Naval Medical Research Center, Silver Spring, MD, USA
| | - Nada Marjanovic
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Clare M Miller
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sagie Mofsowitz
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Venugopalan D Nair
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Edgar Nunez
- Naval Medical Research Center, Silver Spring, MD, USA
| | - Danielle M Parent
- Department of Pathology & Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Chad K Porter
- Naval Medical Research Center, Silver Spring, MD, USA
| | | | | | - Daniel Stadlbauer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Michael Termini
- and Naval Medical Readiness and Training Command Beaufort, Beaufort, SC, USA
| | - Peifang Sun
- Naval Medical Research Center, Silver Spring, MD, USA
| | - Russell P Tracy
- Department of Pathology & Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander Bukreyev
- Department of Pathology University of Texas Medical Branch and Galveston National Laboratory, Galveston, TX, USA
| | - Irene Ramos
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stuart C Sealfon
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
694
|
Saini SK, Hersby DS, Tamhane T, Povlsen HR, Amaya Hernandez SP, Nielsen M, Gang AO, Hadrup SR. SARS-CoV-2 genome-wide T cell epitope mapping reveals immunodominance and substantial CD8 + T cell activation in COVID-19 patients. Sci Immunol 2021; 6:eabf7550. [PMID: 33853928 PMCID: PMC8139428 DOI: 10.1126/sciimmunol.abf7550] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 04/08/2021] [Indexed: 12/11/2022]
Abstract
T cells are important for effective viral clearance, elimination of virus-infected cells and long-term disease protection. To examine the full-spectrum of CD8+ T cell immunity in COVID-19, we experimentally evaluated 3141 major histocompatibility (MHC) class I-binding peptides covering the complete SARS-CoV-2 genome. Using DNA-barcoded peptide-MHC complex (pMHC) multimers combined with a T cell phenotype panel, we report a comprehensive list of 122 immunogenic and a subset of immunodominant SARS-CoV-2 T cell epitopes. Substantial CD8+ T cell recognition was observed in COVID-19 patients, with up to 27% of all CD8+ lymphocytes interacting with SARS-CoV-2-derived epitopes. Most immunogenic regions were derived from open reading frame (ORF) 1 and ORF3, with ORF1 containing most of the immunodominant epitopes. CD8+ T cell recognition of lower affinity was also observed in healthy donors toward SARS-CoV-2-derived epitopes. This pre-existing T cell recognition signature was partially overlapping with the epitope landscape observed in COVID-19 patients and may drive the further expansion of T cell responses to SARS-CoV-2 infection. Importantly the phenotype of the SARS-CoV-2-specific CD8+ T cells, revealed a strong T cell activation in COVID-19 patients, while minimal T cell activation was seen in healthy individuals. We found that patients with severe disease displayed significantly larger SARS-CoV-2-specific T cell populations compared to patients with mild diseases and these T cells displayed a robust activation profile. These results further our understanding of T cell immunity to SARS-CoV-2 infection and hypothesize that strong antigen-specific T cell responses are associated with different disease outcomes.
Collapse
Affiliation(s)
- Sunil Kumar Saini
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark.
| | - Ditte Stampe Hersby
- Department of Haematology, Herlev Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Tripti Tamhane
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Helle Rus Povlsen
- Department of Health Technology, Section of Bioinformatics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Susana Patricia Amaya Hernandez
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Morten Nielsen
- Department of Health Technology, Section of Bioinformatics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Anne Ortved Gang
- Department of Haematology, Herlev Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Sine Reker Hadrup
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark.
| |
Collapse
|
695
|
Yu S, An J, Liao X, Wang H, Ma F, Li D, Li A, Liu W, Zhang S, Liao M, Liu L, Zhao J, Xing S, Wei L, Zhang Z. Distinct kinetics of immunoglobulin isotypes reveal early diagnosis and disease severity of COVID-19: A 6-month follow-up. Clin Transl Med 2021; 11:e342. [PMID: 33784011 PMCID: PMC7989708 DOI: 10.1002/ctm2.342] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 02/04/2021] [Accepted: 02/07/2021] [Indexed: 12/23/2022] Open
Affiliation(s)
- Siyang Yu
- Institute of Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China.,Microbiology Department, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jianghong An
- Institute of Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xuejiao Liao
- Institute of Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Haiyan Wang
- Institute of Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Fen Ma
- Microbiology Department, Harbin Medical University, Harbin, Heilongjiang, China
| | - Dapeng Li
- Institute of Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Aimin Li
- Center Lab of Longhua Branch, Shenzhen People's Hospital, Shenzhen, Guangdong, China
| | - Weilong Liu
- Institute of Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Siwei Zhang
- Microbiology Department, Harbin Medical University, Harbin, Heilongjiang, China
| | - Mingfeng Liao
- Institute of Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Lei Liu
- Institute of Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Juanjuan Zhao
- Institute of Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Shaojun Xing
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen University Health Science Center, Shenzhen, China
| | - Lanlan Wei
- Institute of Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China.,Microbiology Department, Harbin Medical University, Harbin, Heilongjiang, China
| | - Zheng Zhang
- Institute of Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China.,Shenzhen Research Center for Communicable Disease Diagnosis and Treatment of Chinese Academy of Medical Science, Shenzhen, Guangdong, China
| |
Collapse
|
696
|
An efficient system to generate truncated human angiotensin converting enzyme 2 (hACE2) capable of binding RBD and spike protein of SARS-CoV2. Protein Expr Purif 2021; 184:105889. [PMID: 33852951 PMCID: PMC8036248 DOI: 10.1016/j.pep.2021.105889] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/02/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023]
Abstract
Human angiotensin converting enzyme 2 (hACE2) mediates the cell entry of both SARS-CoV and SARS-CoV2 and can be used as a drug target. The DNA encoding the truncated hACE2 (30-356aa) was cloned into pET-28a (+) and expressed in Escherichia coli Rosetta (DE3). The recombinant hACE2 (rhACE2) was purified by affinity chromatography on a Ni-NTA column and characterized with SDS-PAGE and Western blot. The binding activity of rhACE2 to Spike protein of SARS-CoV2 was evaluated in S protein-overexpressed HEK293A cells (HEK293A-SP cells) through flow cytometry. The prokaryotic expression system is able to produce approximately 75 mg protein per liter, which would be useful for infection mechanism study, and drug screening and development of SARS-CoV2.
Collapse
|
697
|
Aziz NA, Corman VM, Echterhoff AKC, Müller MA, Richter A, Schmandke A, Schmidt ML, Schmidt TH, de Vries FM, Drosten C, Breteler MMB. Seroprevalence and correlates of SARS-CoV-2 neutralizing antibodies from a population-based study in Bonn, Germany. Nat Commun 2021; 12:2117. [PMID: 33837204 PMCID: PMC8035181 DOI: 10.1038/s41467-021-22351-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 03/05/2021] [Indexed: 02/07/2023] Open
Abstract
To estimate the seroprevalence and temporal course of SARS-CoV-2 neutralizing antibodies, we embedded a multi-tiered seroprevalence survey within an ongoing community-based cohort study in Bonn, Germany. We first assessed anti-SARS-CoV-2 immunoglobulin G levels with an immunoassay, followed by confirmatory testing of borderline and positive test results with a recombinant spike-based immunofluorescence assay and a plaque reduction neutralization test (PRNT). Those with a borderline or positive immunoassay result were retested after 4 to 5 months. At baseline, 4771 persons participated (88% response rate). Between April 24th and June 30th, 2020, seroprevalence was 0.97% (95% CI: 0.72-1.30) by immunoassay and 0.36% (95% CI: 0.21-0.61) when considering only those with two additional positive confirmatory tests. Importantly, about 20% of PRNT+ individuals lost their neutralizing antibodies within five months. Here, we show that neutralizing antibodies are detectable in only one third of those with a positive immunoassay result, and wane relatively quickly.
Collapse
Affiliation(s)
- N Ahmad Aziz
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, Faculty of Medicine, University of Bonn, Bonn, Germany
| | - Victor M Corman
- Institute of Virology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
- German Center for Infection Research (DZIF), Berlin, Germany
| | - Antje K C Echterhoff
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Marcel A Müller
- Institute of Virology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
- German Center for Infection Research (DZIF), Berlin, Germany
| | - Anja Richter
- Institute of Virology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Antonio Schmandke
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Marie Luisa Schmidt
- Institute of Virology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
- German Center for Infection Research (DZIF), Berlin, Germany
| | - Thomas H Schmidt
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Folgerdiena M de Vries
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Christian Drosten
- Institute of Virology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
- German Center for Infection Research (DZIF), Berlin, Germany
| | - Monique M B Breteler
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
- Institute for Medical Biometry, Informatics and Epidemiology (IMBIE), Faculty of Medicine, University of Bonn, Bonn, Germany.
| |
Collapse
|
698
|
Chirmule N, Nair P, Desai B, Khare R, Nerurkar V, Gaur A. Predicting the severity of disease progression in COVID-19 at the individual and population level: A mathematical model. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.04.01.21254804. [PMID: 33851191 PMCID: PMC8043488 DOI: 10.1101/2021.04.01.21254804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The impact of COVID-19 disease on health and economy has been global, and the magnitude of devastation is unparalleled in modern history. Any potential course of action to manage this complex disease requires the systematic and efficient analysis of data that can delineate the underlying pathogenesis. We have developed a mathematical model of disease progression to predict the clinical outcome, utilizing a set of causal factors known to contribute to COVID-19 pathology such as age, comorbidities, and certain viral and immunological parameters. Viral load and selected indicators of a dysfunctional immune response, such as cytokines IL-6 and IFNα, which contribute to the cytokine storm and fever, parameters of inflammation d-dimer and ferritin, aberrations in lymphocyte number, lymphopenia, and neutralizing antibodies were included for the analysis. The model provides a framework to unravel the multi-factorial complexities of the immune response manifested in SARS-CoV-2 infected individuals. Further, this model can be valuable to predict clinical outcome at an individual level, and to develop strategies for allocating appropriate resources to mitigate severe cases at a population level.
Collapse
Affiliation(s)
| | | | - Bela Desai
- NanoCellect Biomedical, Inc., San Diego, California, USA
| | | | - Vivek Nerurkar
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Amitabh Gaur
- Innovative Assay Solutions LLC, San Diego, California, USA
| |
Collapse
|
699
|
Harrington WE, Trakhimets O, Andrade DV, Dambrauskas N, Raappana A, Jiang Y, Houck J, Selman W, Yang A, Vigdorovich V, Yeung W, Haglund M, Wallner J, Oldroyd A, Hardy S, Stewart SWA, Gervassi A, Van Voorhis W, Frenkel L, Sather DN. Rapid decline of neutralizing antibodies is associated with decay of IgM in adults recovered from mild COVID-19. CELL REPORTS MEDICINE 2021; 2:100253. [PMID: 33842901 PMCID: PMC8020863 DOI: 10.1016/j.xcrm.2021.100253] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/11/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022]
Abstract
The fate of protective immunity following mild severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infection remains ill defined. Here, we characterize antibody responses in a cohort of participants recovered from mild SARS-CoV-2 infection with follow-up to 6 months. We measure immunoglobulin A (IgA), IgM, and IgG binding and avidity to viral antigens and assess neutralizing antibody responses over time. Furthermore, we correlate the effect of fever, gender, age, and time since symptom onset with antibody responses. We observe that total anti-S trimer, anti-receptor-binding domain (RBD), and anti-nucleocapsid protein (NP) IgG are relatively stable over 6 months of follow-up, that anti-S and anti-RBD avidity increases over time, and that fever is associated with higher levels of antibodies. However, neutralizing antibody responses rapidly decay and are strongly associated with declines in IgM levels. Thus, while total antibody against SARS-CoV-2 may persist, functional antibody, particularly IgM, is rapidly lost. These observations have implications for the duration of protective immunity following mild SARS-CoV-2 infection. After mild COVID-19, anti-S-trimer, RBD, and NP IgG are stable for up to 6 months Neutralization activity against the virus rapidly decays over time Neutralization is most strongly correlated with anti-S-trimer IgM titers Antibodies are initially higher in those with fever but reach similar nadirs
Collapse
Affiliation(s)
- Whitney E Harrington
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA.,Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Olesya Trakhimets
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Daniela V Andrade
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Nicholas Dambrauskas
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Andrew Raappana
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Yonghou Jiang
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - John Houck
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - William Selman
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Ashton Yang
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Vladimir Vigdorovich
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Winnie Yeung
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Micaela Haglund
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Jackson Wallner
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Alyssa Oldroyd
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Samantha Hardy
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Samuel W A Stewart
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Ana Gervassi
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Wes Van Voorhis
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA 98109, USA
| | - Lisa Frenkel
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA.,Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - D Noah Sather
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA.,Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
700
|
Velikova T. INFECTION-ACQUIRED VERSUS VACCINE-INDUCED IMMUNITY AGAINST COVID-19. CENTRAL ASIAN JOURNAL OF MEDICAL HYPOTHESES AND ETHICS 2021. [DOI: 10.47316/cajmhe.2021.2.1.05] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The course of COVID-19 depends on a dynamic interplay between SARS-CoV-2 and the host's immune system. Although it is an emerging global health issue, little is known about the specificity, safety, and duration of the immunity elicited by the virus. This hypothesis article explores the benefits of infection-acquired and vaccine-induced immunity against COVID-19, suggesting that the latter outweighs the former. Comparative studies are proposed to explain and reveal all aspects of the immune responses. Although vaccine development relies on studies of naturally acquired immune responses, there are still no comparative analyses of the natural and vaccine immunity against SARS-CoV-2. Moreover, there are scarce reports on the characteristics of both types of responses. The scientific facts about the virulence of SARS-CoV-2 affecting the immune system are of great importance for proposed comparative analyses. Various immunological methods can be employed to elucidate infection-acquired and vaccine-induced immunity against SARS-CoV-2. The safe vaccination of subjects with and without COVID-19 history may disrupt the virus spreading and end the pandemic.
Collapse
|