1
|
Djaïleb A, Parker MF, Lavallée É, Stuible M, Durocher Y, Thériault M, Santerre K, Gilbert C, Boudreau D, Baz M, Masson JF, Langlois MA, Trottier S, Quaglia D, Pelletier JN. Longitudinal determination of seroprevalence and immune response to SARS-CoV-2 in a population of food and retail workers through decentralized testing and transformation of ELISA datasets. PLoS One 2024; 19:e0314499. [PMID: 39680559 DOI: 10.1371/journal.pone.0314499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/11/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Since the onset of the global COVID-19 pandemic in early 2020, numerous studies have been conducted worldwide to understand our immune response to the virus and to vaccination. This study investigates the humoral response elicited by SARS-CoV-2 infection and by vaccination in the poorly studied population of food and retail workers. These occupations were classified as essential by the Public Health Agency of Canada, potentially placing this population at greater risk of infection. Such a risk requires access to reliable and adaptable serological assays that can be rapidly deployed to guide public health strategies. Here we investigate the benefits and limitations of applying adaptable, decentralized tests for population-level immune surveillance in response to a pandemic, even before centralized testing is available. METHODS AND FINDINGS The 1.5-year study period spans from early 2021, when vaccination became available in this region, to mid-2022, following the emergence of the first Omicron variants. The cohort of 304 food and retail workers was recruited in the Québec City area. Participants attended five evenly spaced visits, providing blood samples as well as information on SARS-CoV-2 symptoms or risk factors, prior antigen or PCR test results and vaccination status, as well as work-related risk factors and protective measures. Parallel COVID-19 serological assays were performed using both a standardized chemiluminescent ELISA assay at the centralized platform operated in partnership with the Public Health Agency of Canada, and a semi-automated in-house colorimetric ELISA assay developed at our decentralized site. The YES/NO determination of SARS-CoV-2 vaccine seroconversion and/or infection events using the SARS-CoV-2 ancestral spike protein and nucleocapsid protein validated coherence of the centralized and decentralized assays. The flexibility of the decentralized assays allowed broadening the study to determine cross-reactivity of IgG directed against the spike protein of the SARS-CoV-2 Delta and Omicron VOCs, and IgM directed against the ancestral spike and nucleocapsid proteins. The nature of the data obtained in the decentralized assays allowed treatment with a recently developed mathematical transformation to obtain normal distribution, enabling ANOVA-Welsh statistical analysis. Although no significant differences were observed in humoral response as related to BMI, age, level of education, or chronic illnesses in this cohort of workers, statistically higher levels of vaccine-induced antibodies were observed for restaurant workers and hardware store workers in the early stages of the study, compared to workers in bars and grocery stores and in non-smokers versus smokers. CONCLUSIONS This work highlights the importance of developing adaptable, decentralized tests for population-level immune surveillance in response to a pandemic, even before centralized testing is available. To our knowledge, no other study has reported such an extensive longitudinal investigation during key periods of the COVID-19 pandemic in a cohort of food and retail workers to analyze two types of immunoglobulin, three epitopes and antigens to three VOC. This study will inform strategies and measures to be implemented in the event of a future pandemic.
Collapse
Affiliation(s)
- Abdelhadi Djaïleb
- Département de Chimie, Université de Montréal, Montréal, Canada
- PROTEO, Regroupement Québécois de Recherche sur la Fonction, L'Ingénierie et les Applications des Protéines, Québec, Canada
- Centre en Chimie Verte et Catalyse, Université de Montréal, Montréal, Canada
| | - Megan-Faye Parker
- PROTEO, Regroupement Québécois de Recherche sur la Fonction, L'Ingénierie et les Applications des Protéines, Québec, Canada
- Centre en Chimie Verte et Catalyse, Université de Montréal, Montréal, Canada
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, Canada
| | - Étienne Lavallée
- Département de Chimie, Université de Montréal, Montréal, Canada
- PROTEO, Regroupement Québécois de Recherche sur la Fonction, L'Ingénierie et les Applications des Protéines, Québec, Canada
- Centre en Chimie Verte et Catalyse, Université de Montréal, Montréal, Canada
| | - Matthew Stuible
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Canada
| | - Yves Durocher
- PROTEO, Regroupement Québécois de Recherche sur la Fonction, L'Ingénierie et les Applications des Protéines, Québec, Canada
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Canada
| | - Mathieu Thériault
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, Université Laval, Québec, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Université Laval, Québec, Canada
| | - Kim Santerre
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, Université Laval, Québec, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Université Laval, Québec, Canada
| | - Caroline Gilbert
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, Université Laval, Québec, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Université Laval, Québec, Canada
| | - Denis Boudreau
- Département de Chimie, Université Laval, Québec, Canada
- Centre d'Optique, Photonique et Laser, Université Laval, Québec, Canada
| | - Mariana Baz
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, Université Laval, Québec, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Université Laval, Québec, Canada
| | - Jean-Francois Masson
- Département de Chimie, Université de Montréal, Montréal, Canada
- Institut Courtois, Université de Montréal, Montréal, Canada
- Centre Québécois sur les Matériaux Fonctionnels, Regroupement Québécois sur les Matériaux de Pointe, Centre Interdisciplinaire de Recherche sur le Cerveau et l'Apprentissage, Montréal, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Ottawa Center for Infection, Immunity and Inflammation (CI3), Ottawa, Canada
| | - Sylvie Trottier
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, Université Laval, Québec, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Université Laval, Québec, Canada
| | - Daniela Quaglia
- Département de Chimie, Université de Montréal, Montréal, Canada
- PROTEO, Regroupement Québécois de Recherche sur la Fonction, L'Ingénierie et les Applications des Protéines, Québec, Canada
- Centre en Chimie Verte et Catalyse, Université de Montréal, Montréal, Canada
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, Canada
- Département de Chimie, Université du Québec à Montréal, Montréal, Canada
| | - Joelle N Pelletier
- Département de Chimie, Université de Montréal, Montréal, Canada
- PROTEO, Regroupement Québécois de Recherche sur la Fonction, L'Ingénierie et les Applications des Protéines, Québec, Canada
- Centre en Chimie Verte et Catalyse, Université de Montréal, Montréal, Canada
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, Canada
| |
Collapse
|
2
|
Lugo-Trampe A, López-Cifuentes D, Mendoza-Pérez P, Tafurt-Cardona Y, Joo-Domínguez ADJ, Rios-Ibarra CP, Espinoza-Ruiz M, Chang-Rueda C, Rodriguez-Sanchez IP, Martinez-Fierro ML, Delgado-Enciso I, Trujillo-Murillo KDC. Nine-Month Trend of IgG Antibody Persistence and Associated Symptoms Post-SARS-CoV-2 Infection. Healthcare (Basel) 2024; 12:948. [PMID: 38727505 PMCID: PMC11083704 DOI: 10.3390/healthcare12090948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Between 2 and 8.5% of patients who recover from COVID-19 do not develop antibodies, and the durability of IgG antibodies is under scrutiny. Therefore, the presence and persistence of IgM and IgG antibodies were evaluated in a group of patients diagnosed with SARS-CoV-2 from May to August 2020. Out of 2199 suspected COVID-19 cases, 1264 were confirmed for SARS-CoV-2 by rRT-PCR; 328 consented to participate in the study, with 220 participants followed for 9 months, including 124 men (56%) and 96 women (44%). The primary symptoms were headache, dry cough, and fever. IgG antibodies developed in 95% of patients within 4 weeks post-diagnosis, and a second evaluation at 9 months showed that 72.7% still had detectable IgG antibodies. The presence of IgM in one individual (0.45%) suggested the possibility of reinfection.
Collapse
Affiliation(s)
- Angel Lugo-Trampe
- Faculty of Human Medicine, Campus IV, Universidad Autónoma de Chiapas, Tapachula 30700, Mexico
- Genodiagnóstica SA de CV, Tapachula, Chiapas 30700, Mexico
| | - Daniel López-Cifuentes
- Faculty of Chemistry Sciences, Campus IV, Universidad Autónoma de Chiapas, Tapachula 30700, Mexico
| | - Paúl Mendoza-Pérez
- Faculty of Human Medicine, Campus IV, Universidad Autónoma de Chiapas, Tapachula 30700, Mexico
| | - Yaliana Tafurt-Cardona
- Faculty of Human Medicine, Campus IV, Universidad Autónoma de Chiapas, Tapachula 30700, Mexico
| | | | - Clara Patricia Rios-Ibarra
- Medical and Pharmaceutical Biotechnology Unit, Center for Research and Assistance in Technology and Design of the State of Jalisco (CIATEJ), Guadalajara 44270, Mexico
| | - Marisol Espinoza-Ruiz
- Faculty of Chemistry Sciences, Campus IV, Universidad Autónoma de Chiapas, Tapachula 30700, Mexico
| | - Consuelo Chang-Rueda
- Faculty of Chemistry Sciences, Campus IV, Universidad Autónoma de Chiapas, Tapachula 30700, Mexico
| | - Iram Pablo Rodriguez-Sanchez
- Molecular and Structural Physiology Laboratory, School of Biological Sciences, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66455, Mexico
| | - Margarita L. Martinez-Fierro
- Molecular Medicine Laboratory, Unidad de Medicina Humana y Ciencias de la Salud, Universidad Autónoma de Zacatecas, Zacatecas 98160, Mexico
| | - Iván Delgado-Enciso
- School of Medicine, University of Colima, Colima 28040, Mexico
- Colima Cancerology State Institute, IMSS-Bienestar, Colima 28085, Mexico
| | | |
Collapse
|
3
|
Peterhoff D, Wiegrebe S, Einhauser S, Patt AJ, Beileke S, Günther F, Steininger P, Niller HH, Burkhardt R, Küchenhoff H, Gefeller O, Überla K, Heid IM, Wagner R. Population-based study of the durability of humoral immunity after SARS-CoV-2 infection. Front Immunol 2023; 14:1242536. [PMID: 37868969 PMCID: PMC10585261 DOI: 10.3389/fimmu.2023.1242536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 09/19/2023] [Indexed: 10/24/2023] Open
Abstract
SARS-CoV-2 antibody quantity and quality are key markers of humoral immunity. However, there is substantial uncertainty about their durability. We investigated levels and temporal change of SARS-CoV-2 antibody quantity and quality. We analyzed sera (8 binding, 4 avidity assays for spike-(S-)protein and nucleocapsid-(N-)protein; neutralization) from 211 seropositive unvaccinated participants, from the population-based longitudinal TiKoCo study, at three time points within one year after infection with the ancestral SARS-CoV-2 virus. We found a significant decline of neutralization titers and binding antibody levels in most assays (linear mixed regression model, p<0.01). S-specific serum avidity increased markedly over time, in contrast to N-specific. Binding antibody levels were higher in older versus younger participants - a difference that disappeared for the asymptomatic-infected. We found stronger antibody decline in men versus women and lower binding and avidity levels in current versus never-smokers. Our comprehensive longitudinal analyses across 13 antibody assays suggest decreased neutralization-based protection and prolonged affinity maturation within one year after infection.
Collapse
Affiliation(s)
- David Peterhoff
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Simon Wiegrebe
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
- Statistical Consulting Unit StaBLab, Department of Statistics, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Sebastian Einhauser
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
| | - Arisha J. Patt
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Stephanie Beileke
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Felix Günther
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
- Statistical Consulting Unit StaBLab, Department of Statistics, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Philipp Steininger
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Hans H. Niller
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Helmut Küchenhoff
- Statistical Consulting Unit StaBLab, Department of Statistics, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Olaf Gefeller
- Department of Medical Informatics, Biometry and Epidemiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Klaus Überla
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Iris M. Heid
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
4
|
Batistela CM, Correa DPF, Bueno ÁM, Piqueira JRC. SIRSi-vaccine dynamical model for the Covid-19 pandemic. ISA TRANSACTIONS 2023; 139:391-405. [PMID: 37217378 PMCID: PMC10186248 DOI: 10.1016/j.isatra.2023.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 04/17/2023] [Accepted: 05/12/2023] [Indexed: 05/24/2023]
Abstract
Covid-19, caused by severe acute respiratory syndrome coronavirus 2, broke out as a pandemic during the beginning of 2020. The rapid spread of the disease prompted an unprecedented global response involving academic institutions, regulatory agencies, and industries. Vaccination and nonpharmaceutical interventions including social distancing have proven to be the most effective strategies to combat the pandemic. In this context, it is crucial to understand the dynamic behavior of the Covid-19 spread together with possible vaccination strategies. In this study, a susceptible-infected-removed-sick model with vaccination (SIRSi-vaccine) was proposed, accounting for the unreported yet infectious. The model considered the possibility of temporary immunity following infection or vaccination. Both situations contribute toward the spread of diseases. The transcritical bifurcation diagram of alternating and mutually exclusive stabilities for both disease-free and endemic equilibria were determined in the parameter space of vaccination rate and isolation index. The existing equilibrium conditions for both points were determined in terms of the epidemiological parameters of the model. The bifurcation diagram allowed us to estimate the maximum number of confirmed cases expected for each set of parameters. The model was fitted with data from São Paulo, the state capital of SP, Brazil, which describes the number of confirmed infected cases and the isolation index for the considered data window. Furthermore, simulation results demonstrate the possibility of periodic undamped oscillatory behavior of the susceptible population and the number of confirmed cases forced by the periodic small-amplitude oscillations in the isolation index. The main contributions of the proposed model are as follows: A minimum effort was required when vaccination was combined with social isolation, while additionally ensuring the existence of equilibrium points. The model could provide valuable information for policymakers, helping define disease prevention mitigation strategies that combine vaccination and non-pharmaceutical interventions, such as social distancing and the use of masks. In addition, the SIRSi-vaccine model facilitated the qualitative assessment of information regarding the unreported infected yet infectious cases, while considering temporary immunity, vaccination, and social isolation index.
Collapse
Affiliation(s)
| | - Diego P F Correa
- Federal University of ABC - UFABC - São Bernardo do Campo, SP, Brazil.
| | - Átila M Bueno
- Polytechnic School of University of São Paulo, São Paulo, SP, Brazil.
| | | |
Collapse
|
5
|
Li Z, Xiang T, Liang B, Liu J, Deng H, Yang X, Wang H, Feng X, Zelinskyy G, Trilling M, Sutter K, Lu M, Dittmer U, Wang B, Yang D, Zheng X, Liu J. SARS-CoV-2-specific T cell responses wane profoundly in convalescent individuals 10 months after primary infection. Virol Sin 2023; 38:606-619. [PMID: 37414153 PMCID: PMC10436107 DOI: 10.1016/j.virs.2023.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 06/28/2023] [Indexed: 07/08/2023] Open
Abstract
A key question in the coronavirus disease 2019 (COVID-19) pandemic is the duration of specific T cell responses against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) post primary infection, which is difficult to address due to the large-scale COVID-19 vaccination and re-exposure to the virus. Here, we conducted an analysis of the long-term SARS-CoV-2-specific T cell responses in a unique cohort of convalescent individuals (CIs) that were among the first to be infected worldwide and without any possible antigen re-exposure since then. The magnitude and breadth of SARS-CoV-2-specific T cell responses correlated inversely with the time that had elapsed from disease onset and the age of those CIs. The mean magnitude of SARS-CoV-2-specific CD4 and CD8 T cell responses decreased about 82% and 76%, respectively, over the time period of ten months after infection. Accordingly, the longitudinal analysis also demonstrated that SARS-CoV-2-specific T cell responses waned significantly in 75% of CIs during the follow-up. Collectively, we provide a comprehensive characterization of the long-term memory T cell response in CIs, suggesting that robust SARS-CoV-2-specific T cell immunity post primary infection may be less durable than previously expected.
Collapse
Affiliation(s)
- Ziwei Li
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tiandan Xiang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Boyun Liang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Liu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hui Deng
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xuecheng Yang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hua Wang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xuemei Feng
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Gennadiy Zelinskyy
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, 45147, Germany; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Mirko Trilling
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, 45147, Germany; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kathrin Sutter
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, 45147, Germany; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Mengji Lu
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, 45147, Germany; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ulf Dittmer
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, 45147, Germany; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Baoju Wang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Dongliang Yang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Xin Zheng
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Jia Liu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
6
|
Goenka MK, Goenka U, Patil VU, Das SS, Afzalpurkar S, Jajodia S, Mukherjee M, Shah BB, Moitra S. Kinetics of Covid-19 antibodies in terms of titre and duration among healthcare workers: A longitudinal study. THE NATIONAL MEDICAL JOURNAL OF INDIA 2023; 35:201-205. [PMID: 36715043 DOI: 10.25259/nmji_109_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Background Most individuals with Covid-19 infection develop antibodies specific to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, the dynamics of these antibodies is variable and not well-studied. We aimed to determine the titres of naturally acquired antibodies over a 12-week follow-up. Methods We recruited healthcare workers who had tested positive on a specific quantitative reverse transcription-polymerase chain reaction (qRT-PCR) for SARS-CoV-2, and then tested for the presence of immunoglobulin G (IgG) antibody against the same virus at baseline and again at 6 and 12 weeks. The antibody titre was determined by a semi-quantitative assay based on signal/cut-off ratio. Healthcare workers with antibody positivity were divided into those with high titre (ratio ≥12) and low titre (<12). Their demographic details and risk factors were surveyed through a Google form and analysed in relation to the antibody titres at three time-points. Results Of the 286 healthcare workers, 10.48% had high antibody titres. Healthcare workers who had tested positive by qRT-PCR and those who had received the Bacille Calmette-Guérin (BCG) vaccination or other immune-boosters had a higher frequency of high antibody titres. While there was a significant decline in antibody titres at 6 and 12 weeks, 87.46% of individuals positive for IgG antibody persisted to have the antibody even at 12 weeks. Conclusion Healthcare workers who tested positive for SARS-CoV-2 on qRT-PCR had a high positivity for the specific antibody, which continued to express in them even at 12 weeks. Further follow-up is likely to enhance our understanding of antibody kinetics following SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Mahesh Kumar Goenka
- Department of Clinical Imaging and Interventional Radiology, Apollo Multispeciality Hospitals, Institute of Gastrosciences and Liver, Kolkata, West Bengal, India
| | - Usha Goenka
- Department of Clinical Imaging and Interventional Radiology, Apollo Multispeciality Hospitals, Institute of Gastrosciences and Liver, Kolkata, West Bengal, India
| | - Vikram Uttam Patil
- Department of Clinical Imaging and Interventional Radiology, Apollo Multispeciality Hospitals, Institute of Gastrosciences and Liver, Kolkata, West Bengal, India
| | - Sudipta Sekhar Das
- Department of Transfusion Medicine and Blood Bank, Apollo Multispeciality Hospitals, Institute of Gastrosciences and Liver, Kolkata, West Bengal, India
| | - Shivaraj Afzalpurkar
- Department of Clinical Imaging and Interventional Radiology, Apollo Multispeciality Hospitals, Institute of Gastrosciences and Liver, Kolkata, West Bengal, India
| | - Surabhi Jajodia
- Department of Clinical Imaging and Interventional Radiology, Apollo Multispeciality Hospitals, Institute of Gastrosciences and Liver, Kolkata, West Bengal, India
| | - Muhuya Mukherjee
- Department of Biostatistics, Apollo Multispeciality Hospitals, Institute of Gastrosciences and Liver, Kolkata, West Bengal, India
| | - Bhavik Bharat Shah
- Department of Clinical Imaging and Interventional Radiology, Apollo Multispeciality Hospitals, Institute of Gastrosciences and Liver, Kolkata, West Bengal, India
| | - Saibal Moitra
- Department of Allergy and Asthma Research Centre, Apollo Multispeciality Hospitals, Institute of Gastrosciences and Liver, Kolkata, West Bengal, India
| |
Collapse
|
7
|
De Thoisy A, Woudenberg T, Pelleau S, Donnadieu F, Garcia L, Pinaud L, Tondeur L, Meola A, Arowas L, Clement N, Backovic M, Ungeheuer MN, Fontanet A, White M. Seroepidemiology of the Seasonal Human Coronaviruses NL63, 229E, OC43 and HKU1 in France. Open Forum Infect Dis 2023; 10:ofad340. [PMID: 37496603 PMCID: PMC10368309 DOI: 10.1093/ofid/ofad340] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/30/2023] [Indexed: 07/28/2023] Open
Abstract
Background The seasonal human coronaviruses (HCoV) NL63, 229E, OC43, and HKU1 are globally endemic, yet the majority of HCoV infections remain undiagnosed. Methods In a cross-sectional study, 2389 serum samples were collected from children and adults in France in 2020. In a longitudinal cohort study, 2520 samples were collected from 898 French individuals followed up between 2020 and 2021. Antibodies to HCoVs were measured using a bead-based multiplex assay. Results The rate of waning of anti-HCoV spike immunoglobulin G antibodies was estimated as 0.22-0.47 year-1 for children, and 0.13-0.27 year-1 for adults. Seroreversion was estimated as 0.31-1.37 year-1 in children and 0.19-0.72 year-1 in adults. The estimated seroconversion rate in children was consistent with 20%-39% of children being infected every year with each HCoV. Conclusions The high force of infection in children indicates that HCoVs may be responsible for a substantial proportion of fever episodes experienced by children.
Collapse
Affiliation(s)
- Alix De Thoisy
- Infectious Disease Epidemiology and Analytics G5 Unit, Department of Global Health, Institut Pasteur, Université Paris Cité, Paris, France
| | - Tom Woudenberg
- Infectious Disease Epidemiology and Analytics G5 Unit, Department of Global Health, Institut Pasteur, Université Paris Cité, Paris, France
| | - Stéphane Pelleau
- Correspondence: Michael White, PhD, Infectious Disease Epidemiology and Analytics G5 Unit, Department of Global Health, Institut Pasteur, Rue du Docteur Roux, Paris 75015, France (); Stéphane Pelleau, PhD, Infectious Disease Epidemiology and Analytics G5 Unit, Department of Global Health, Institut Pasteur, Rue du Docteur Roux, Paris 75015, France ()
| | - Françoise Donnadieu
- Infectious Disease Epidemiology and Analytics G5 Unit, Department of Global Health, Institut Pasteur, Université Paris Cité, Paris, France
| | - Laura Garcia
- Infectious Disease Epidemiology and Analytics G5 Unit, Department of Global Health, Institut Pasteur, Université Paris Cité, Paris, France
| | - Laurie Pinaud
- Epidemiology of Emerging Diseases Unit, Department of Global Health, Institut Pasteur, Université Paris Cité, Paris, France
| | - Laura Tondeur
- Epidemiology of Emerging Diseases Unit, Department of Global Health, Institut Pasteur, Université Paris Cité, Paris, France
| | - Annalisa Meola
- Structural Virology Unit, Department of Virology and CNRS UMR 3569, Institut Pasteur, Université Paris Cité, Paris, France
| | - Laurence Arowas
- Investigation Clinique et Accès aux Ressources Biologiques (ICAReB), Center for Translational Research, Institut Pasteur, Paris, France
| | - Nathalie Clement
- Coordination Clinique du CRT, Center for Translational Research, Institut Pasteur, Paris, France
| | - Marija Backovic
- Structural Virology Unit, Department of Virology and CNRS UMR 3569, Institut Pasteur, Université Paris Cité, Paris, France
| | - Marie-Noëlle Ungeheuer
- Investigation Clinique et Accès aux Ressources Biologiques (ICAReB), Center for Translational Research, Institut Pasteur, Paris, France
| | - Arnaud Fontanet
- Epidemiology of Emerging Diseases Unit, Department of Global Health, Institut Pasteur, Université Paris Cité, Paris, France
- PACRI Unit, Conservatoire National des Arts et Métiers, Paris, France
| | - Michael White
- Correspondence: Michael White, PhD, Infectious Disease Epidemiology and Analytics G5 Unit, Department of Global Health, Institut Pasteur, Rue du Docteur Roux, Paris 75015, France (); Stéphane Pelleau, PhD, Infectious Disease Epidemiology and Analytics G5 Unit, Department of Global Health, Institut Pasteur, Rue du Docteur Roux, Paris 75015, France ()
| | | |
Collapse
|
8
|
Iannizzi C, Chai KL, Piechotta V, Valk SJ, Kimber C, Monsef I, Wood EM, Lamikanra AA, Roberts DJ, McQuilten Z, So-Osman C, Jindal A, Cryns N, Estcourt LJ, Kreuzberger N, Skoetz N. Convalescent plasma for people with COVID-19: a living systematic review. Cochrane Database Syst Rev 2023; 5:CD013600. [PMID: 37162745 PMCID: PMC10171886 DOI: 10.1002/14651858.cd013600.pub6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
BACKGROUND Convalescent plasma may reduce mortality in patients with viral respiratory diseases, and is being investigated as a potential therapy for coronavirus disease 2019 (COVID-19). A thorough understanding of the current body of evidence regarding benefits and risks of this intervention is required. OBJECTIVES To assess the effectiveness and safety of convalescent plasma transfusion in the treatment of people with COVID-19; and to maintain the currency of the evidence using a living systematic review approach. SEARCH METHODS To identify completed and ongoing studies, we searched the World Health Organization (WHO) COVID-19 Global literature on coronavirus disease Research Database, MEDLINE, Embase, Cochrane COVID-19 Study Register, and the Epistemonikos COVID-19 L*OVE Platform. We searched monthly until 03 March 2022. SELECTION CRITERIA We included randomised controlled trials (RCTs) evaluating convalescent plasma for COVID-19, irrespective of disease severity, age, gender or ethnicity. We excluded studies that included populations with other coronavirus diseases (severe acute respiratory syndrome (SARS) or Middle East respiratory syndrome (MERS)), as well as studies evaluating standard immunoglobulin. DATA COLLECTION AND ANALYSIS We followed standard Cochrane methodology. To assess bias in included studies we used RoB 2. We used the GRADE approach to rate the certainty of evidence for the following outcomes: all-cause mortality at up to day 28, worsening and improvement of clinical status (for individuals with moderate to severe disease), hospital admission or death, COVID-19 symptoms resolution (for individuals with mild disease), quality of life, grade 3 or 4 adverse events, and serious adverse events. MAIN RESULTS In this fourth review update version, we included 33 RCTs with 24,861 participants, of whom 11,432 received convalescent plasma. Of these, nine studies are single-centre studies and 24 are multi-centre studies. Fourteen studies took place in America, eight in Europe, three in South-East Asia, two in Africa, two in western Pacific and three in eastern Mediterranean regions and one in multiple regions. We identified a further 49 ongoing studies evaluating convalescent plasma, and 33 studies reporting as being completed. Individuals with a confirmed diagnosis of COVID-19 and moderate to severe disease 29 RCTs investigated the use of convalescent plasma for 22,728 participants with moderate to severe disease. 23 RCTs with 22,020 participants compared convalescent plasma to placebo or standard care alone, five compared to standard plasma and one compared to human immunoglobulin. We evaluate subgroups on detection of antibodies detection, symptom onset, country income groups and several co-morbidities in the full text. Convalescent plasma versus placebo or standard care alone Convalescent plasma does not reduce all-cause mortality at up to day 28 (risk ratio (RR) 0.98, 95% confidence interval (CI) 0.92 to 1.03; 220 per 1000; 21 RCTs, 19,021 participants; high-certainty evidence). It has little to no impact on need for invasive mechanical ventilation, or death (RR 1.03, 95% CI 0.97 to 1.11; 296 per 1000; 6 RCTs, 14,477 participants; high-certainty evidence) and has no impact on whether participants are discharged from hospital (RR 1.00, 95% CI 0.97 to 1.02; 665 per 1000; 6 RCTs, 12,721 participants; high-certainty evidence). Convalescent plasma may have little to no impact on quality of life (MD 1.00, 95% CI -2.14 to 4.14; 1 RCT, 483 participants; low-certainty evidence). Convalescent plasma may have little to no impact on the risk of grades 3 and 4 adverse events (RR 1.17, 95% CI 0.96 to 1.42; 212 per 1000; 6 RCTs, 2392 participants; low-certainty evidence). It has probably little to no effect on the risk of serious adverse events (RR 1.14, 95% CI 0.91 to 1.44; 135 per 1000; 6 RCTs, 3901 participants; moderate-certainty evidence). Convalescent plasma versus standard plasma We are uncertain whether convalescent plasma reduces or increases all-cause mortality at up to day 28 (RR 0.73, 95% CI 0.45 to 1.19; 129 per 1000; 4 RCTs, 484 participants; very low-certainty evidence). We are uncertain whether convalescent plasma reduces or increases the need for invasive mechanical ventilation, or death (RR 5.59, 95% CI 0.29 to 108.38; 311 per 1000; 1 study, 34 participants; very low-certainty evidence) and whether it reduces or increases the risk of serious adverse events (RR 0.80, 95% CI 0.55 to 1.15; 236 per 1000; 3 RCTs, 327 participants; very low-certainty evidence). We did not identify any study reporting other key outcomes. Convalescent plasma versus human immunoglobulin Convalescent plasma may have little to no effect on all-cause mortality at up to day 28 (RR 1.07, 95% CI 0.76 to 1.50; 464 per 1000; 1 study, 190 participants; low-certainty evidence). We did not identify any study reporting other key outcomes. Individuals with a confirmed diagnosis of SARS-CoV-2 infection and mild disease We identified two RCTs reporting on 536 participants, comparing convalescent plasma to placebo or standard care alone, and two RCTs reporting on 1597 participants with mild disease, comparing convalescent plasma to standard plasma. Convalescent plasma versus placebo or standard care alone We are uncertain whether convalescent plasma reduces all-cause mortality at up to day 28 (odds ratio (OR) 0.36, 95% CI 0.09 to 1.46; 8 per 1000; 2 RCTs, 536 participants; very low-certainty evidence). It may have little to no effect on admission to hospital or death within 28 days (RR 1.05, 95% CI 0.60 to 1.84; 117 per 1000; 1 RCT, 376 participants; low-certainty evidence), on time to COVID-19 symptom resolution (hazard ratio (HR) 1.05, 95% CI 0.85 to 1.30; 483 per 1000; 1 RCT, 376 participants; low-certainty evidence), on the risk of grades 3 and 4 adverse events (RR 1.29, 95% CI 0.75 to 2.19; 144 per 1000; 1 RCT, 376 participants; low-certainty evidence) and the risk of serious adverse events (RR 1.14, 95% CI 0.66 to 1.94; 133 per 1000; 1 RCT, 376 participants; low-certainty evidence). We did not identify any study reporting other key outcomes. Convalescent plasma versus standard plasma We are uncertain whether convalescent plasma reduces all-cause mortality at up to day 28 (OR 0.30, 95% CI 0.05 to 1.75; 2 per 1000; 2 RCTs, 1597 participants; very low-certainty evidence). It probably reduces admission to hospital or death within 28 days (RR 0.49, 95% CI 0.31 to 0.75; 36 per 1000; 2 RCTs, 1595 participants; moderate-certainty evidence). Convalescent plasma may have little to no effect on initial symptom resolution at up to day 28 (RR 1.12, 95% CI 0.98 to 1.27; 1 RCT, 416 participants; low-certainty evidence). We did not identify any study reporting other key outcomes. This is a living systematic review. We search monthly for new evidence and update the review when we identify relevant new evidence. AUTHORS' CONCLUSIONS For the comparison of convalescent plasma versus placebo or standard care alone, our certainty in the evidence that convalescent plasma for individuals with moderate to severe disease does not reduce mortality and has little to no impact on clinical improvement or worsening is high. It probably has little to no effect on SAEs. For individuals with mild disease, we have very-low to low certainty evidence for most primary outcomes and moderate certainty for hospital admission or death. There are 49 ongoing studies, and 33 studies reported as complete in a trials registry. Publication of ongoing studies might resolve some of the uncertainties around convalescent plasma therapy for people with asymptomatic or mild disease.
Collapse
Affiliation(s)
- Claire Iannizzi
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Khai Li Chai
- Transfusion Research Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Vanessa Piechotta
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Sarah J Valk
- Jon J van Rood Center for Clinical Transfusion Research, Sanquin/Leiden University Medical Center, Leiden, Netherlands
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Catherine Kimber
- Systematic Review Initiative, NHS Blood and Transplant, Oxford, UK
| | - Ina Monsef
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Erica M Wood
- Transfusion Research Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | | | - David J Roberts
- Systematic Review Initiative, NHS Blood and Transplant, Oxford, UK
| | - Zoe McQuilten
- Transfusion Research Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Cynthia So-Osman
- Sanquin Blood Bank, Amsterdam, Netherlands
- Erasmus Medical Centre, Rotterdam, Netherlands
| | - Aikaj Jindal
- Department of Transfusion Medicine, SPS Hospitals, Ludhiana (Punjab), India
| | - Nora Cryns
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Lise J Estcourt
- Haematology/Transfusion Medicine, NHS Blood and Transplant, Oxford, UK
| | - Nina Kreuzberger
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Nicole Skoetz
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
9
|
Monitoring of SARS-CoV-2 Infection in Ragusa Area: Next Generation Sequencing and Serological Analysis. Int J Mol Sci 2023; 24:ijms24054742. [PMID: 36902172 PMCID: PMC10003428 DOI: 10.3390/ijms24054742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
The coronavirus disease 19 (COVID-19) post pandemic evolution is correlated to the development of new variants. Viral genomic and immune response monitoring are fundamental to the surveillance of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Since 1 January to 31 July 2022, we monitored the SARS-CoV-2 variants trend in Ragusa area sequencing n.600 samples by next generation sequencing (NGS) technology: n.300 were healthcare workers (HCWs) of ASP Ragusa. The evaluation of anti-Nucleocapside (N), receptor-binding domain (RBD), the two subunit of S protein (S1 and S2) IgG levels in 300 exposed vs. 300 unexposed HCWs to SARS-CoV-2 was performed. Differences in immune response and clinical symptoms related to the different variants were investigated. The SARS-CoV-2 variants trend in Ragusa area and in Sicily region were comparable. BA.1 and BA.2 were the most representative variants, whereas the diffusion of BA.3 and BA.4 affected some places of the region. Although no correlation was found between variants and clinical manifestations, anti-N and anti-S2 levels were positively correlated with an increase in the symptoms number. SARS-CoV-2 infection induced a statistically significant enhancement in antibody titers compared to that produced by SARS-CoV-2 vaccine administration. In post-pandemic period, the evaluation of anti-N IgG could be used as an early marker to identify asymptomatic subjects.
Collapse
|
10
|
Wachman EM, Snyder-Cappione J, Devera J, Boateng J, Dhole Y, Clarke K, Yuen RR, Parker SE, Hunnewell J, Ferraro R, Jean-Sicard S, Woodard E, Cruikshank A, Sinha B, Bartolome R, Barnett ED, Yarrington C, Taglauer ES, Sabharwal V. Maternal, Infant, and Breast Milk Antibody Response Following COVID-19 Infection in Early Versus Late Gestation. Pediatr Infect Dis J 2023; 42:e70-e76. [PMID: 36729773 PMCID: PMC9935237 DOI: 10.1097/inf.0000000000003802] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/29/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND Coronavirus disease 2019 [severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)] infection at varying time points during the pregnancy can influence antibody levels after delivery. We aimed to examine SARS-CoV-2 IgG, IgM and IgA receptor binding domain of the spike protein and nucleocapsid protein (N-protein) reactive antibody concentrations in maternal blood, infant blood and breastmilk at birth and 6 weeks after SARS-CoV-2 infection in early versus late gestation. METHODS Mothers with SARS-CoV-2 infection during pregnancy were enrolled between July 2020 and May 2021. Maternal blood, infant blood and breast milk samples were collected at delivery and 6 weeks postpartum. Samples were analyzed for SARS-CoV-2 spike and N-protein reactive IgG, IgM and IgA antibodies. Antibody concentrations were compared at the 2 time points and based on trimester of infection ("early" 1st/2nd vs. "late" 3rd). RESULTS Dyads from 20 early and 11 late trimester infections were analyzed. For the entire cohort, there were no significant differences in antibody levels at delivery versus 6 weeks with the exception of breast milk levels which declined over time. Early gestation infections were associated with higher levels of breastmilk IgA to spike protein ( P = 0.04). Infant IgG levels to spike protein were higher at 6 weeks after late infections ( P = 0.04). There were strong correlations between maternal and infant IgG levels at delivery ( P < 0.01), and between breastmilk and infant IgG levels. CONCLUSIONS SARS-CoV-2 infection in early versus late gestation leads to a persistent antibody response in maternal blood, infant blood and breast milk over the first 6 weeks after delivery.
Collapse
Affiliation(s)
- Elisha M. Wachman
- From the Department of Pediatrics, Boston Medical Center, Boston, Massachusetts
| | | | - Jean Devera
- Boston University School of Medicine, Boston, Massachusetts
| | - Jeffery Boateng
- From the Department of Pediatrics, Boston Medical Center, Boston, Massachusetts
| | - Yashoda Dhole
- Boston University School of Medicine, Boston, Massachusetts
| | - Katherine Clarke
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts
| | - Rachel R. Yuen
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts
| | - Samantha E. Parker
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts
| | - Jessica Hunnewell
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts
| | - Renee Ferraro
- From the Department of Pediatrics, Boston Medical Center, Boston, Massachusetts
| | - Sigride Jean-Sicard
- From the Department of Pediatrics, Boston Medical Center, Boston, Massachusetts
| | - Elizabeth Woodard
- From the Department of Pediatrics, Boston Medical Center, Boston, Massachusetts
| | - Alice Cruikshank
- From the Department of Pediatrics, Boston Medical Center, Boston, Massachusetts
| | - Bharati Sinha
- From the Department of Pediatrics, Boston Medical Center, Boston, Massachusetts
| | - Ruby Bartolome
- From the Department of Pediatrics, Boston Medical Center, Boston, Massachusetts
| | | | - Christina Yarrington
- Department of Obstetrics and Gynecology, Boston Medical Center, Boston, Massachusetts
| | | | - Vishakha Sabharwal
- From the Department of Pediatrics, Boston Medical Center, Boston, Massachusetts
| |
Collapse
|
11
|
Iannizzi C, Chai KL, Piechotta V, Valk SJ, Kimber C, Monsef I, Wood EM, Lamikanra AA, Roberts DJ, McQuilten Z, So-Osman C, Jindal A, Cryns N, Estcourt LJ, Kreuzberger N, Skoetz N. Convalescent plasma for people with COVID-19: a living systematic review. Cochrane Database Syst Rev 2023; 2:CD013600. [PMID: 36734509 PMCID: PMC9891348 DOI: 10.1002/14651858.cd013600.pub5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Convalescent plasma may reduce mortality in patients with viral respiratory diseases, and is being investigated as a potential therapy for coronavirus disease 2019 (COVID-19). A thorough understanding of the current body of evidence regarding benefits and risks of this intervention is required. OBJECTIVES To assess the effectiveness and safety of convalescent plasma transfusion in the treatment of people with COVID-19; and to maintain the currency of the evidence using a living systematic review approach. SEARCH METHODS To identify completed and ongoing studies, we searched the World Health Organization (WHO) COVID-19 Global literature on coronavirus disease Research Database, MEDLINE, Embase, Cochrane COVID-19 Study Register, and the Epistemonikos COVID-19 L*OVE Platform. We searched monthly until 03 March 2022. SELECTION CRITERIA We included randomised controlled trials (RCTs) evaluating convalescent plasma for COVID-19, irrespective of disease severity, age, gender or ethnicity. We excluded studies that included populations with other coronavirus diseases (severe acute respiratory syndrome (SARS) or Middle East respiratory syndrome (MERS)), as well as studies evaluating standard immunoglobulin. DATA COLLECTION AND ANALYSIS We followed standard Cochrane methodology. To assess bias in included studies we used RoB 2. We used the GRADE approach to rate the certainty of evidence for the following outcomes: all-cause mortality at up to day 28, worsening and improvement of clinical status (for individuals with moderate to severe disease), hospital admission or death, COVID-19 symptoms resolution (for individuals with mild disease), quality of life, grade 3 or 4 adverse events, and serious adverse events. MAIN RESULTS In this fourth review update version, we included 33 RCTs with 24,861 participants, of whom 11,432 received convalescent plasma. Of these, nine studies are single-centre studies and 24 are multi-centre studies. Fourteen studies took place in America, eight in Europe, three in South-East Asia, two in Africa, two in western Pacific and three in eastern Mediterranean regions and one in multiple regions. We identified a further 49 ongoing studies evaluating convalescent plasma, and 33 studies reporting as being completed. Individuals with a confirmed diagnosis of COVID-19 and moderate to severe disease 29 RCTs investigated the use of convalescent plasma for 22,728 participants with moderate to severe disease. 23 RCTs with 22,020 participants compared convalescent plasma to placebo or standard care alone, five compared to standard plasma and one compared to human immunoglobulin. We evaluate subgroups on detection of antibodies detection, symptom onset, country income groups and several co-morbidities in the full text. Convalescent plasma versus placebo or standard care alone Convalescent plasma does not reduce all-cause mortality at up to day 28 (risk ratio (RR) 0.98, 95% confidence interval (CI) 0.92 to 1.03; 220 per 1000; 21 RCTs, 19,021 participants; high-certainty evidence). It has little to no impact on need for invasive mechanical ventilation, or death (RR 1.03, 95% CI 0.97 to 1.11; 296 per 1000; 6 RCTs, 14,477 participants; high-certainty evidence) and has no impact on whether participants are discharged from hospital (RR 1.00, 95% CI 0.97 to 1.02; 665 per 1000; 6 RCTs, 12,721 participants; high-certainty evidence). Convalescent plasma may have little to no impact on quality of life (MD 1.00, 95% CI -2.14 to 4.14; 1 RCT, 483 participants; low-certainty evidence). Convalescent plasma may have little to no impact on the risk of grades 3 and 4 adverse events (RR 1.17, 95% CI 0.96 to 1.42; 212 per 1000; 6 RCTs, 2392 participants; low-certainty evidence). It has probably little to no effect on the risk of serious adverse events (RR 1.14, 95% CI 0.91 to 1.44; 135 per 1000; 6 RCTs, 3901 participants; moderate-certainty evidence). Convalescent plasma versus standard plasma We are uncertain whether convalescent plasma reduces or increases all-cause mortality at up to day 28 (RR 0.73, 95% CI 0.45 to 1.19; 129 per 1000; 4 RCTs, 484 participants; very low-certainty evidence). We are uncertain whether convalescent plasma reduces or increases the need for invasive mechanical ventilation, or death (RR 5.59, 95% CI 0.29 to 108.38; 311 per 1000; 1 study, 34 participants; very low-certainty evidence) and whether it reduces or increases the risk of serious adverse events (RR 0.80, 95% CI 0.55 to 1.15; 236 per 1000; 3 RCTs, 327 participants; very low-certainty evidence). We did not identify any study reporting other key outcomes. Convalescent plasma versus human immunoglobulin Convalescent plasma may have little to no effect on all-cause mortality at up to day 28 (RR 1.07, 95% CI 0.76 to 1.50; 464 per 1000; 1 study, 190 participants; low-certainty evidence). We did not identify any study reporting other key outcomes. Individuals with a confirmed diagnosis of SARS-CoV-2 infection and mild disease We identified two RCTs reporting on 536 participants, comparing convalescent plasma to placebo or standard care alone, and two RCTs reporting on 1597 participants with mild disease, comparing convalescent plasma to standard plasma. Convalescent plasma versus placebo or standard care alone We are uncertain whether convalescent plasma reduces all-cause mortality at up to day 28 (odds ratio (OR) 0.36, 95% CI 0.09 to 1.46; 8 per 1000; 2 RCTs, 536 participants; very low-certainty evidence). It may have little to no effect on admission to hospital or death within 28 days (RR 1.05, 95% CI 0.60 to 1.84; 117 per 1000; 1 RCT, 376 participants; low-certainty evidence), on time to COVID-19 symptom resolution (hazard ratio (HR) 1.05, 95% CI 0.85 to 1.30; 483 per 1000; 1 RCT, 376 participants; low-certainty evidence), on the risk of grades 3 and 4 adverse events (RR 1.29, 95% CI 0.75 to 2.19; 144 per 1000; 1 RCT, 376 participants; low-certainty evidence) and the risk of serious adverse events (RR 1.14, 95% CI 0.66 to 1.94; 133 per 1000; 1 RCT, 376 participants; low-certainty evidence). We did not identify any study reporting other key outcomes. Convalescent plasma versus standard plasma We are uncertain whether convalescent plasma reduces all-cause mortality at up to day 28 (OR 0.30, 95% CI 0.05 to 1.75; 2 per 1000; 2 RCTs, 1597 participants; very low-certainty evidence). It probably reduces admission to hospital or death within 28 days (RR 0.49, 95% CI 0.31 to 0.75; 36 per 1000; 2 RCTs, 1595 participants; moderate-certainty evidence). Convalescent plasma may have little to no effect on initial symptom resolution at up to day 28 (RR 1.12, 95% CI 0.98 to 1.27; 1 RCT, 416 participants; low-certainty evidence). We did not identify any study reporting other key outcomes. This is a living systematic review. We search monthly for new evidence and update the review when we identify relevant new evidence. AUTHORS' CONCLUSIONS For the comparison of convalescent plasma versus placebo or standard care alone, our certainty in the evidence that convalescent plasma for individuals with moderate to severe disease does not reduce mortality and has little to no impact on clinical improvement or worsening is high. It probably has little to no effect on SAEs. For individuals with mild disease, we have low certainty evidence for our primary outcomes. There are 49 ongoing studies, and 33 studies reported as complete in a trials registry. Publication of ongoing studies might resolve some of the uncertainties around convalescent plasma therapy for people with asymptomatic or mild disease.
Collapse
Affiliation(s)
- Claire Iannizzi
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Khai Li Chai
- Transfusion Research Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Vanessa Piechotta
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Sarah J Valk
- Jon J van Rood Center for Clinical Transfusion Research, Sanquin/Leiden University Medical Center, Leiden, Netherlands
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Catherine Kimber
- Systematic Review Initiative, NHS Blood and Transplant, Oxford, UK
| | - Ina Monsef
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Erica M Wood
- Transfusion Research Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | | | - David J Roberts
- Systematic Review Initiative, NHS Blood and Transplant, Oxford, UK
| | - Zoe McQuilten
- Transfusion Research Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Cynthia So-Osman
- Sanquin Blood Bank, Amsterdam, Netherlands
- Erasmus Medical Centre, Rotterdam, Netherlands
| | - Aikaj Jindal
- Department of Transfusion Medicine, SPS Hospitals, Ludhiana (Punjab), India
| | - Nora Cryns
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Lise J Estcourt
- Haematology/Transfusion Medicine, NHS Blood and Transplant, Oxford, UK
| | - Nina Kreuzberger
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Nicole Skoetz
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
12
|
Kimber C, Valk SJ, Chai KL, Piechotta V, Iannizzi C, Monsef I, Wood EM, Lamikanra AA, Roberts DJ, McQuilten Z, So-Osman C, Estcourt LJ, Skoetz N. Hyperimmune immunoglobulin for people with COVID-19. Cochrane Database Syst Rev 2023; 1:CD015167. [PMID: 36700518 PMCID: PMC9887673 DOI: 10.1002/14651858.cd015167.pub2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Hyperimmune immunoglobulin (hIVIG) contains polyclonal antibodies, which can be prepared from large amounts of pooled convalescent plasma or prepared from animal sources through immunisation. They are being investigated as a potential therapy for coronavirus disease 2019 (COVID-19). This review was previously part of a parent review addressing convalescent plasma and hIVIG for people with COVID-19 and was split to address hIVIG and convalescent plasma separately. OBJECTIVES To assess the benefits and harms of hIVIG therapy for the treatment of people with COVID-19, and to maintain the currency of the evidence using a living systematic review approach. SEARCH METHODS To identify completed and ongoing studies, we searched the World Health Organization (WHO) COVID-19 Research Database, the Cochrane COVID-19 Study Register, the Epistemonikos COVID-19 L*OVE Platform and Medline and Embase from 1 January 2019 onwards. We carried out searches on 31 March 2022. SELECTION CRITERIA We included randomised controlled trials (RCTs) that evaluated hIVIG for COVID-19, irrespective of disease severity, age, gender or ethnicity. We excluded studies that included populations with other coronavirus diseases (severe acute respiratory syndrome (SARS) or Middle East respiratory syndrome (MERS)), as well as studies that evaluated standard immunoglobulin. DATA COLLECTION AND ANALYSIS We followed standard Cochrane methodology. To assess bias in included studies, we used RoB 2. We rated the certainty of evidence, using the GRADE approach, for the following outcomes: all-cause mortality, improvement and worsening of clinical status (for individuals with moderate to severe disease), quality of life, adverse events, and serious adverse events. MAIN RESULTS We included five RCTs with 947 participants, of whom 688 received hIVIG prepared from humans, 18 received heterologous swine glyco-humanised polyclonal antibody, and 241 received equine-derived processed and purified F(ab')2 fragments. All participants were hospitalised with moderate-to-severe disease, most participants were not vaccinated (only 12 participants were vaccinated). The studies were conducted before or during the emergence of several SARS-CoV-2 variants of concern. There are no data for people with COVID-19 with no symptoms (asymptomatic) or people with mild COVID-19. We identified a further 10 ongoing studies evaluating hIVIG. Benefits of hIVIG prepared from humans We included data on one RCT (579 participants) that assessed the benefits and harms of hIVIG 0.4 g/kg compared to saline placebo. hIVIG may have little to no impact on all-cause mortality at 28 days (risk ratio (RR) 0.79, 95% confidence interval (CI) 0.43 to 1.44; absolute effect 77 per 1000 with placebo versus 61 per 1000 (33 to 111) with hIVIG; low-certainty evidence). The evidence is very uncertain about the effect on worsening of clinical status at day 7 (RR 0.85, 95% CI 0.58 to 1.23; very low-certainty evidence). It probably has little to no impact on improvement of clinical status on day 28 (RR 1.02, 95% CI 0.97 to 1.08; moderate-certainty evidence). We did not identify any studies that reported quality-of-life outcomes, so we do not know if hIVIG has any impact on quality of life. Harms of hIVIG prepared from humans hIVIG may have little to no impact on adverse events at any grade on day 1 (RR 0.98, 95% CI 0.81 to 1.18; 431 per 1000; 1 study 579 participants; low-certainty evidence). Patients receiving hIVIG probably experience more adverse events at grade 3-4 severity than patients who receive placebo (RR 4.09, 95% CI 1.39 to 12.01; moderate-certainty evidence). hIVIG may have little to no impact on the composite outcome of serious adverse events or death up to day 28 (RR 0.72, 95% CI 0.45 to 1.14; moderate-certainty evidence). We also identified additional results on the benefits and harms of other dose ranges of hIVIG, not included in the summary of findings table, but summarised in additional tables. Benefits of animal-derived polyclonal antibodies We included data on one RCT (241 participants) to assess the benefits and harms of receptor-binding domain-specific polyclonal F(ab´)2 fragments of equine antibodies (EpAbs) compared to saline placebo. EpAbs may reduce all-cause mortality at 28 days (RR 0.60, 95% CI 0.26 to 1.37; absolute effect 114 per 1000 with placebo versus 68 per 1000 (30 to 156) ; low-certainty evidence). EpAbs may reduce worsening of clinical status up to day 28 (RR 0.67, 95% CI 0.38 to 1.18; absolute effect 203 per 1000 with placebo versus 136 per 1000 (77 to 240); low-certainty evidence). It may have some effect on improvement of clinical status on day 28 (RR 1.06, 95% CI 0.96 to 1.17; low-certainty evidence). We did not identify any studies that reported quality-of-life outcomes, so we do not know if EpAbs have any impact on quality of life. Harms of animal-derived polyclonal antibodies EpAbs may have little to no impact on the number of adverse events at any grade up to 28 days (RR 0.99, 95% CI 0.74 to 1.31; low-certainty evidence). Adverse events at grade 3-4 severity were not reported. Individuals receiving EpAbs may experience fewer serious adverse events than patients receiving placebo (RR 0.67, 95% CI 0.38 to 1.19; low-certainty evidence). We also identified additional results on the benefits and harms of other animal-derived polyclonal antibody doses, not included in the summary of findings table, but summarised in additional tables. AUTHORS' CONCLUSIONS We included data from five RCTs that evaluated hIVIG compared to standard therapy, with participants with moderate-to-severe disease. As the studies evaluated different preparations (from humans or from various animals) and doses, we could not pool them. hIVIG prepared from humans may have little to no impact on mortality, and clinical improvement and worsening. hIVIG may increase grade 3-4 adverse events. Studies did not evaluate quality of life. RBD-specific polyclonal F(ab´)2 fragments of equine antibodies may reduce mortality and serious adverse events, and may reduce clinical worsening. However, the studies were conducted before or during the emergence of several SARS-CoV-2 variants of concern and prior to widespread vaccine rollout. As no studies evaluated hIVIG for participants with asymptomatic infection or mild disease, benefits for these individuals remains uncertain. This is a living systematic review. We search monthly for new evidence and update the review when we identify relevant new evidence.
Collapse
Affiliation(s)
- Catherine Kimber
- Systematic Review Initiative, NHS Blood and Transplant, Oxford, UK
| | - Sarah J Valk
- Jon J van Rood Center for Clinical Transfusion Research, Sanquin/Leiden University Medical Center, Leiden, Netherlands
| | - Khai Li Chai
- Transfusion Research Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Vanessa Piechotta
- Cochrane Haematology, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Claire Iannizzi
- Cochrane Haematology, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ina Monsef
- Cochrane Haematology, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Erica M Wood
- Transfusion Research Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | | | - David J Roberts
- Systematic Review Initiative, NHS Blood and Transplant, Oxford, UK
| | - Zoe McQuilten
- Transfusion Research Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Cynthia So-Osman
- Erasmus Medical Centre, Rotterdam, Netherlands
- Unit Transfusion Medicine, Sanquin Blood Supply Foundation, Amsterdam, Netherlands
| | - Lise J Estcourt
- Haematology/Transfusion Medicine, NHS Blood and Transplant, Oxford, UK
| | - Nicole Skoetz
- Cochrane Haematology, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
13
|
Assaid N, Arich S, Charoute H, Akarid K, Anouar Sadat M, Maaroufi A, Ezzikouri S, Sarih M. Kinetics of SARS-CoV-2 IgM and IgG Antibodies 3 Months after COVID-19 Onset in Moroccan Patients. Am J Trop Med Hyg 2023; 108:145-154. [PMID: 36509045 PMCID: PMC9833093 DOI: 10.4269/ajtmh.22-0448] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 09/24/2022] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses serious global public health problems. Characterization of the immune response, particularly antibodies to SARS-CoV-2, is important for establishing vaccine strategies. The purpose of this study was to evaluate longitudinally the kinetics of anti-SARS-CoV-2 antibodies against spike protein (S1) for up to 3 months in a cohort of 169 COVID-19 patients. We enrolled COVID-19 patients at two regional hospitals in Casablanca, Morocco, between March and September 2021. Blood samples were collected and N-specific IgM and S-specific IgG levels were measured by a commercial Euroimmun ELISA. IgM antibodies were assessed 2-5 (D00), 9-12 (D07), 17-20 (D15), and 32-37 (D30) days after symptom onset; IgG antibodies were assessed at these time points plus 60 (D60) and 90 (D90) days after symptom onset. We found that at 3 months after symptom onset, 79% of patients had detectable SARS-CoV-2-specific IgG antibodies, whereas their IgM seropositivity was 19% by 1 month after symptom onset. The IgM level decreased to 0.34 (interquartile range [IQR] 0.19-0.92) at 1 month after symptom onset, whereas the IgG level peaked at D30 (3.10; IQR 1.83-5.64) and remained almost stable at D90 (2.95; IQR 1.52-5.19). IgG levels were significantly higher in patients older than 50 years than in those younger than 50 at all follow-up time points (P < 0.05). Statistical analysis showed no significant difference in median anti-S1 antibody levels among infected patients based on gender or comorbidities. This study provides information on the longevity of anti-SARS-CoV-2 IgM and IgG antibodies in COVID-19 patients.
Collapse
Affiliation(s)
- Najlaa Assaid
- Service de Parasitologie et des Maladies Vectorielles, Institut Pasteur du Maroc, Casablanca, Morocco;,Biochemistry, Biotechnology and Immunophysiopathology Research Team, Health and Environment Laboratory, Aïn Chock Faculty of Sciences, University of Hassan II Casablanca, Casablanca, Morocco
| | - Soukaina Arich
- Service de Parasitologie et des Maladies Vectorielles, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Hicham Charoute
- Research Unit of Epidemiology, Biostatistics and Bioinformatics, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Khadija Akarid
- Biochemistry, Biotechnology and Immunophysiopathology Research Team, Health and Environment Laboratory, Aïn Chock Faculty of Sciences, University of Hassan II Casablanca, Casablanca, Morocco
| | - Mohamed Anouar Sadat
- Research Unit of Epidemiology, Biostatistics and Bioinformatics, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Abderrahmane Maaroufi
- Service de Parasitologie et des Maladies Vectorielles, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Sayeh Ezzikouri
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - M’hammed Sarih
- Service de Parasitologie et des Maladies Vectorielles, Institut Pasteur du Maroc, Casablanca, Morocco;,Address correspondence to M’hammed Sarih, Service de Parasitologie et des Maladies Vectorielles, Institut Pasteur du Maroc, Place Louis Pasteur, Casablanca, Morocco. E-mail:
| |
Collapse
|
14
|
Dowran R, Damavandi AR, Azad TM. Reinfection and reactivation of SARS-CoV-2. Future Virol 2022. [PMID: 36176508 PMCID: PMC9514089 DOI: 10.2217/fvl-2021-0212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 08/31/2022] [Indexed: 11/21/2022]
Abstract
As the cases of SARS-CoV-2 infection escalates, the essence of in-depth knowledge around acquired immunity and emergence of reinfection and reactivation have to be captured. While being a rare phenomenon, reinfection occurs as the result of diminishing protection conferred by antibodies, especially IgG. Reactivation is more concerned with the role of various elements including shedding lingering viral RNA for a prolonged time and incomplete resolution of infection along with the insight of dormant viral exosomes’ role. The concept of testing positive after two consecutive negative results requires proper discrimination of reinfection from reactivation. In this review, we summarized the current evidence for possible mechanisms leading to viral reactivation or test re-positivity. We also pointed out risk factors associated with both reinfection and reactivation.
Collapse
Affiliation(s)
- Razieh Dowran
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.,Student Scientific Association of Virology, Student Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirmasoud Rayati Damavandi
- Student Scientific Association of Virology, Student Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Talat Mokhtari Azad
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Kinetics of anti-nucleocapsid IgG response in COVID-19 immunocompetent convalescent patients. Sci Rep 2022; 12:12403. [PMID: 35859108 PMCID: PMC9297274 DOI: 10.1038/s41598-022-16402-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 07/11/2022] [Indexed: 01/02/2023] Open
Abstract
The comprehension of a long-term humoral immune response against SARS-CoV-2 can shed light on the treatment and vaccination strategies of COVID-19 disease, improving the knowledge about this virus infection and/or re-infection. We assessed the IgG antibodies against SARS-CoV-2 nucleocapsid (N) protein (anti-SARS-CoV-2 (N) IgG) in 1441 COVID-19 convalescent patients within 15 months longitudinal study from middle-developed country. The main inclusion criteria was positive RT– PCR result on nasopharyngeal swab samples at least one month before antibody testing and absence of any induced or inherited immunodeficiency. 92.7% of convalescent patients’ serum contained anti-SARS-CoV-2 (N) IgG and only 1.3% of patients had a delayed antibody response. In the majority of convalescent patients’ the durability of antibodies lasted more than one year. The kinetics of anti-SARS-CoV-2 (N) IgG took a bell-shaped character—increased first 25–30 weeks, then started to decrease, but were still detectable for more than 15 months. We found that on the one hand anti-SARS-CoV-2 humoral response level correlates with disease severity, on the other, in particular, the level of peak antibodies correlates with age—older patients develop more robust humoral response regardless of sex, disease severity and BMI.
Collapse
|
16
|
Yang J, Ma L, Guo L, Zhang T, Leng Z, Jia M, Chen F, Qi W, Zhang X, Wang Q, Yang Y, Feng L, Ren L, Yang W, Wang C. Seroprevalence and dynamics of anti-SARS-CoV-2 antibodies: a longitudinal study based on patients with underlying diseases in Wuhan. Respir Res 2022; 23:188. [PMID: 35841095 PMCID: PMC9284953 DOI: 10.1186/s12931-022-02096-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 06/22/2022] [Indexed: 12/13/2022] Open
Abstract
Background Assessing the humoral immunity of patients with underlying diseases after being infected with SARS-CoV-2 is essential for adopting effective prevention and control strategies. The purpose of this study is to analyze the seroprevalence of people with underlying diseases and the dynamic change features of anti-SARS-CoV-2 antibodies. Methods We selected 100 communities in Wuhan using the probability-proportional-to-size sampling method. From these 100 communities, we randomly selected households according to a list provided by the local government. Individuals who have lived in Wuhan for at least 14 days since December 2019 and were ≥ 40 years old were included. From April 9–13, 2020, community staff invited all selected individuals to the community healthcare center in batches by going door-to-door or telephone. All participants completed a standardized electronic questionnaire simultaneously. Finally, 5 ml of venous blood was collected from all participants. Blood samples were tested for the presence of pan-immunoglobulins, IgM, IgA, and IgG antibodies against SARS-CoV-2 nucleocapsid protein and neutralising antibodies were assessed. During the period June 11–13, 2020 and October 9–December 5, 2020, all family members of a positive family and matched negative families were followed up twice. Results The seroprevalence of anti-SARS-CoV-2 antibodies in people with underlying diseases was 6.30% (95% CI [5.09–7.52]), and that of people without underlying diseases was 6.12% (95% CI [5.33–6.91]). A total of 313 people were positive for total antibodies at baseline, of which 97 had underlying disease. At the first follow-up, a total of 212 people were positive for total antibodies, of which 66 had underlying disease. At the second follow-up, a total of 238 people were positive for total antibodies, of which 68 had underlying disease. A total of 219 participants had three consecutive serum samples with positive total antibodies at baseline. The IgG titers decreased significantly with or without underlying diseases (P < 0.05) within the 9 months at least, while the neutralizing antibody titer remained stable. The titer of asymptomatic patients was lower than that of symptomatic patients (baseline, P = 0.032, second follow-up, P = 0.018) in the underlying diseases group. Conclusion Our research focused on the serological changes of people with and without underlying diseases in a state of single natural infection. Regardless of the underlying diseases, the IgG titer decreased significantly over time, while there was no significant difference in the decline rate of IgG between with and without underlying diseases. Moreover, the neutralizing antibody titer remained relatively stable within the 9 months at least. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02096-5.
Collapse
Affiliation(s)
- Jin Yang
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Libing Ma
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Institute of Pharmaceutical and Medical Devices Supervision, National Medical Products Administration-Chinese Academy of Medical Sciences, Beijing, China.,Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Li Guo
- National Health Commission Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Respiratory Disease Pathogenomics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ting Zhang
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Institute of Pharmaceutical and Medical Devices Supervision, National Medical Products Administration-Chinese Academy of Medical Sciences, Beijing, China
| | - Zhiwei Leng
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Institute of Pharmaceutical and Medical Devices Supervision, National Medical Products Administration-Chinese Academy of Medical Sciences, Beijing, China
| | - Mengmeng Jia
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Institute of Pharmaceutical and Medical Devices Supervision, National Medical Products Administration-Chinese Academy of Medical Sciences, Beijing, China
| | - Fangyuan Chen
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Institute of Pharmaceutical and Medical Devices Supervision, National Medical Products Administration-Chinese Academy of Medical Sciences, Beijing, China
| | - Weiran Qi
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xingxing Zhang
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing Wang
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan Yang
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Luzhao Feng
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Institute of Pharmaceutical and Medical Devices Supervision, National Medical Products Administration-Chinese Academy of Medical Sciences, Beijing, China
| | - Lili Ren
- National Health Commission Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China. .,Key Laboratory of Respiratory Disease Pathogenomics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Weizhong Yang
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China. .,Institute of Pharmaceutical and Medical Devices Supervision, National Medical Products Administration-Chinese Academy of Medical Sciences, Beijing, China.
| | - Chen Wang
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
17
|
Muslim Dawood S, Khudhur Al Joofy I. Evaluation of IgM and IgG in COVID-19 Recovered Patients in Iraq. ARCHIVES OF RAZI INSTITUTE 2022; 77:1191-1197. [PMID: 36618307 PMCID: PMC9759244 DOI: 10.22092/ari.2022.357515.2054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/01/2022] [Indexed: 01/10/2023]
Abstract
Severe acute respiratory syndrome coronavirus-2 is a major threat to health care worldwide with high morbidity and mortality. Therefore, understanding the role of immune mechanisms and humoral response is vital in this disease. The present study aimed to investigate the relationship between Immunoglobulins (IgM, IgG) in COVID-19 recovered patients with age, gender, and severity of the disease. The duration of effect of antibody levels and protection against re-infection has also been evaluated in the patients. Three groups participated in this study; group 1: 0-14 days after recovery, group 2: 2 months after recovery, group 3: 3 months after recovery, group 4: 4-6 months after recovery, group 5: more than 6 months. The nasopharyngeal swab was used to confirm recovery by Real-Time Polymerase Chain Reaction (RT-PCR) technique. IgM and IgG antibody levels were evaluated using Enzyme-Linked Immuno Fluorescent Assay (ELIFA) technique. The results indicated that the IgM levels increased for one month during the seven days after infection and then decreased in most patients (P≤0.05). The mean of IgG in group 1 increased compared to those of other studied groups. A significant decrease was observed in group 2 compared to group 1, as well as in group 3 compared to groups 1, and 2. Also, a significant difference existed between group 4 compared to groups 1, 2, and 3. Finally, significant differences were noticed between group 5 compared to groups 1, 2, 3, and 4 (P≤0.05). No significant differences were observed in antibodies level between male, and female COVID-19 recovered patients in groups 1, 2, 3, 4, and 5 (P≤0.05). Finally, highly significant differences in IgG levels between mild, moderate, and severe subgroups in groups 1 and 2. The present study demonstrated that IgM and IgG against SARS-CoV-2 appeared in the early stages of the disease and decreased after 1 month and failed to maintain high levels during the 6-month observation.
Collapse
Affiliation(s)
- S Muslim Dawood
- Educational Laboratories, Unit of Clinical Immunology, Baghdad Medical City, Baghdad, Iraq
| | - I Khudhur Al Joofy
- Department of Biology, College of Science, Mustansiriyah University, Baghdad, Iraq
| |
Collapse
|
18
|
Immune response to COVID-19 vaccination in a population with and without a previous SARS-CoV-2 infection. Ir J Med Sci 2022; 192:731-739. [PMID: 35676470 PMCID: PMC9177345 DOI: 10.1007/s11845-022-03044-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/01/2022] [Indexed: 11/01/2022]
Abstract
PURPOSE To evaluate IgG production in a group of vaccinated and unvaccinated subjects previously infected, or not, with SARS-CoV-2. METHODS A total of 316 subjects were enrolled at different times after vaccination and/or infection. IgG against target S1 subunit of the spike protein of SARS-COV-2 was assessed by a chemiluminescent microparticle immunoassay. Participant data was collected using a clinical-epidemiological survey. RESULTS A total of 56.2% (n = 146) of our cohort was vaccinated, with 27.5% (n = 36) reporting a previous infection. Of these, all were IgG positive at the time of the study, regardless of gender, age category, vaccine type, and elapsed time since vaccination. The vaccinated group without a previous infection (72.5%, n = 95) showed a slightly lower IgG seropositivity and median values, overall, although significantly higher in females and lower with the ChAdOx1 nCoV-19 (AstraZeneca) vaccine. Vaccinated subjects above the age of 65 showed a trend towards higher median IgG values (13,911.0 AU/mL), when previously infected with SARS-CoV-2, but comparatively lower IgG median value (5158.7 AU/mL) in its absence. In all vaccinated groups, IgG antibody production increased at 1-2 weeks, peaking at 4-6 weeks. Afterward, IgG decreased progressively but almost all subjects (97.7%, n = 128) were seropositive for the remainder of our study. Fully vaccinated individuals with a past infection showed a lower IgG rate of decrease versus their uninfected counterparts (17.9 vs 22.6%, respectively). CONCLUSION Our findings suggest a higher effect of vaccination on the production IgG antibodies, as opposed to natural infection. Nonetheless, in general, antibody titers waned rapidly.
Collapse
|
19
|
Doke P, Gothankar JS, Doke PP, Kulkarni MM, Khalate KK, Shrivastava S, Patil JR, Arankalle VA. Time dependent decline of neutralizing antibody titers in COVID-19 patients from Pune, India and evidence of reinfection. Microbes Infect 2022; 24:104979. [PMID: 35452812 PMCID: PMC9020493 DOI: 10.1016/j.micinf.2022.104979] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 03/09/2022] [Accepted: 04/07/2022] [Indexed: 01/25/2023]
Abstract
PURPOSE To assess modulation of neutralizing antibody titers in COVID-19 patients and understand association of variables such as age, presence of comorbidity, BMI and gender with antibody titers. METHODS Patients (n = 100) diagnosed from 20th March 2020 to 17th August 2020 and treated at two large hospitals from Pune, India were included and followed up (clinical and serologic) for varied periods. IgG-anti-SARS-CoV-2 (Spike protein-based ELISA) and neutralizing antibody titers (NAb, PRNT) were determined in all the samples. RESULTS Of the 100 patients enrolled initially (median 60 days of diagnosis), follow up samples were collected from 70 patients (median 106 days of diagnosis). Overall, NAb titers reduced significantly (p < 0.001) and as early as 3-4 months. During two visits, 20% and 7.1% patients reported some symptoms. At the first visit, NAb titers were higher in patients with severe disease (p < 0.001), comorbidities (p < 0.005), age <50 years (p < 0.05) and male gender (p < 0.05). Multivariate analysis identified older age (p < 0.001), duration post-diagnosis and female gender as independent variables influencing NAb titers (negative correlation, p < 0.05). During the follow-up, reduction in NAb titers was recorded in patients with comorbidity (p < 0.05), mild disease (p < 0.05), age <50 years (p < 0.05), higher BMI (p < 0.05) and male gender (p < 0.001). Serology identified six cases of asymptomatic reinfections. CONCLUSIONS Decline of NAb titers was associated with age <50 years, mild disease, comorbidities, higher BMI and male gender. At the time of follow up, 8/70 (11.4%) patients lacked neutralizing antibodies. Evidence of 6 probable asymptomatic reinfections suggests waning of immunity, but, probable protection from clinical disease needing hospitalization.
Collapse
Affiliation(s)
- Purwa Doke
- Department of Medicine, Bharati Vidyapeeth Deemed University Medical College, Pune-Satara Road, Katraj, Pune 411043, Maharashtra, India
| | - Jayshree Sachin Gothankar
- Department of Community Medicine, Bharati Vidyapeeth Deemed University Medical College, Pune-Satara Road, Katraj, Pune 411043, Maharashtra, India
| | - Prakash Prabhakarrao Doke
- Department of Community Medicine, Bharati Vidyapeeth Deemed University Medical College, Pune-Satara Road, Katraj, Pune 411043, Maharashtra, India
| | | | | | - Shubham Shrivastava
- Department of Communicable Diseases, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune-Satara Road, Katraj, Pune, Maharashtra, India
| | - Jayesh Rangrao Patil
- Department of Community Medicine, Bharati Vidyapeeth Deemed University Medical College, Pune-Satara Road, Katraj, Pune 411043, Maharashtra, India
| | - Vidya Avinash Arankalle
- Department of Communicable Diseases, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune-Satara Road, Katraj, Pune, Maharashtra, India,Corresponding author
| |
Collapse
|
20
|
Dufour-Gaume F, Javelle E, Sailliol A, Cap AP, Prat NJ. COVID-19 : viral infection, endotheliopathy and the immuno-inflammatory response… is it time to consider a standard (non-immunized) plasma therapy approach to maintain homeostasis? Transfus Clin Biol 2022; 29:191-194. [PMID: 35644840 PMCID: PMC9059338 DOI: 10.1016/j.tracli.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 11/29/2022]
Affiliation(s)
| | - Emilie Javelle
- Hôpital d'Instruction des Armées Laveran, Marseille, France
| | - Anne Sailliol
- Institut de Recherche Biomédicale des Armées, France
| | - Andre P Cap
- U.S. Army Institute of Surgical Research, Houston, USA
| | | |
Collapse
|
21
|
Rando HM, Brueffer C, Lordan R, Dattoli AA, Manheim D, Meyer JG, Mundo AI, Perrin D, Mai D, Wellhausen N, Gitter A, Greene CS. Molecular and Serologic Diagnostic Technologies for SARS-CoV-2. ARXIV 2022:arXiv:2204.12598v2. [PMID: 35547240 PMCID: PMC9094103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Revised: 04/28/2022] [Indexed: 01/09/2023]
Abstract
The COVID-19 pandemic has presented many challenges that have spurred biotechnological research to address specific problems. Diagnostics is one area where biotechnology has been critical. Diagnostic tests play a vital role in managing a viral threat by facilitating the detection of infected and/or recovered individuals. From the perspective of what information is provided, these tests fall into two major categories, molecular and serological. Molecular diagnostic techniques assay whether a virus is present in a biological sample, thus making it possible to identify individuals who are currently infected. Additionally, when the immune system is exposed to a virus, it responds by producing antibodies specific to the virus. Serological tests make it possible to identify individuals who have mounted an immune response to a virus of interest and therefore facilitate the identification of individuals who have previously encountered the virus. These two categories of tests provide different perspectives valuable to understanding the spread of SARS-CoV-2. Within these categories, different biotechnological approaches offer specific advantages and disadvantages. Here we review the categories of tests developed for the detection of the SARS-CoV-2 virus or antibodies against SARS-CoV-2 and discuss the role of diagnostics in the COVID-19 pandemic.
Collapse
Affiliation(s)
- Halie M Rando
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America; Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, United States of America; Center for Health AI, University of Colorado School of Medicine, Aurora, Colorado, United States of America · Funded by the Gordon and Betty Moore Foundation (GBMF 4552); the National Human Genome Research Institute (R01 HG010067)
| | | | - Ronan Lordan
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-5158, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA 19104, USA
| | - Anna Ada Dattoli
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David Manheim
- 1DaySooner, Delaware, United States of America; Risk and Health Communication Research Center, School of Public Health, University of Haifa, Haifa, Israel; Technion, Israel Institute of Technology, Haifa, Israel · Funded by Center for Effective Altruism, Long Term Future Fund
| | - Jesse G Meyer
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America · Funded by National Institute of General Medical Sciences (R35 GM142502)
| | - Ariel I Mundo
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| | - Dimitri Perrin
- School of Computer Science, Queensland University of Technology, Brisbane, Australia; Centre for Data Science, Queensland University of Technology, Brisbane, Australia
| | - David Mai
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, and Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA, USA
| | - Nils Wellhausen
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Anthony Gitter
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America; Morgridge Institute for Research, Madison, Wisconsin, United States of America · Funded by John W. and Jeanne M. Rowe Center for Research in Virology
| | - Casey S Greene
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America; Childhood Cancer Data Lab, Alex's Lemonade Stand Foundation, Philadelphia, Pennsylvania, United States of America; Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, United States of America; Center for Health AI, University of Colorado School of Medicine, Aurora, Colorado, United States of America · Funded by the Gordon and Betty Moore Foundation (GBMF 4552); the National Human Genome Research Institute (R01 HG010067)
| |
Collapse
|
22
|
Kaygusuz S, Korukluoğlu G, Coşgun Y, Şahin Ö, Arslan F. INVESTIGATION AND LONG-TERM MONITORING OF THE PRESENCE OF NEUTRALIZING ANTIBODY IN PATIENTS WITH COVID-19 DISEASE OF DIFFERENT CLINICAL SEVERITY. J Med Virol 2022; 94:3596-3604. [PMID: 35365870 DOI: 10.1002/jmv.27751] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 03/28/2022] [Indexed: 01/08/2023]
Abstract
OBJECTIVES Understanding the immune responses elicited by SARS-CoV-2 infection is critical to public health policy and vaccine development and prevention of reinfections for COVID-19. It is important to know the neutralizing capacity of antibodies and to monitor their persistence. METHODS Patients with Covid-19 were divided into four groups (severe-critical, moderate, mild and asymptomatic) according to their clinical severity. Antibodies against SARS-CoV-2 Spike viral surface protein were investigated by ELISA method 3 months and 9 months after the onset of the disease. Neutralizing antibody (NAb) response was evaluated by microneutralization test. Patients who received at least two doses of COVID-19 vaccine after illness were enrolled. RESULTS SARS-CoV-2 IgG and NAb titers were shown to be strongly correlated with disease severity. Anti-SARS-CoV-2 IgG and neutralizing antibody levels were found to be compatible with each other. After 9 months of follow-up, both IgG and NA levels continued unabated in individuals who had the disease. In individuals who received at least two doses of the vaccine, these levels increased, except for severe-critical patients. CONCLUSIONS High levels of anti-SARS-CoV-2 IgG are indicative, as it is difficult to investigate NAb in routine laboratories. At the same time, it can be predicted that this period may be much longer if it continues for at least 9 months and is reinforced with vaccination. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Sedat Kaygusuz
- Kırıkkale University Faculty of Medicine, Department of Infection Disease and Clinical Microbiology, Kirikkale, Turkey
| | - Gülay Korukluoğlu
- Republic of Turkey Ministry of Health, General Directorate of Public Health, National Virology Reference Laboratory, Ankara, Turkey
| | - Yasemin Coşgun
- Republic of Turkey Ministry of Health, General Directorate of Public Health, National Virology Reference Laboratory, Ankara, Turkey
| | - Ömer Şahin
- Kırıkkale University Faculty of Medicine, Department of Infection Disease and Clinical Microbiology, Kirikkale, Turkey
| | - Ferhat Arslan
- Kırıkkale University Faculty of Medicine, Department of Infection Disease and Clinical Microbiology, Kirikkale, Turkey
| |
Collapse
|
23
|
Abstract
Severe acute respiratory syndrome coronavirus 2 (COVID)-19 has emerged as the greatest global health threat in generations. An unprecedented mobilization of researchers has generated a wealth of data on humoral responses to SARS-CoV-2 within a year of the pandemic's beginning. The rapidly developed understanding of acute-phase antibody induction and medium-term antibody durability in COVID-19 is important at an individual level to inform patient care and a population level to help predict transmission dynamics. In this brief review, we will describe the development and maintenance of antibody responses to immunization and infections generally and the specific antibody dynamics observed for COVID-19. These crucial features of the humoral response have implications for the use of antibody therapeutics against the virus and can inform the likelihood of reinfection of individuals by the virus.
Collapse
Affiliation(s)
- Adam Zuiani
- Department of Medicine, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA,Department of Medicine, Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA,BioNTech, Cambridge, MA 02139, USA
| | - Duane R. Wesemann
- Department of Medicine, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA,Department of Medicine, Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA,Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA,Corresponding author. Department of Medicine, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139
| |
Collapse
|
24
|
Pradenas E, Trinité B, Urrea V, Marfil S, Tarrés-Freixas F, Ortiz R, Rovirosa C, Rodon J, Vergara-Alert J, Segalés J, Guallar V, Valencia A, Izquierdo-Useros N, Noguera-Julian M, Carrillo J, Paredes R, Mateu L, Chamorro A, Toledo R, Massanella M, Clotet B, Blanco J. Clinical course impacts early kinetics,magnitude, and amplitude of SARS-CoV-2 neutralizing antibodies beyond 1 year after infection. Cell Rep Med 2022; 3:100523. [PMID: 35233547 PMCID: PMC8784437 DOI: 10.1016/j.xcrm.2022.100523] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/12/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022]
Abstract
To understand the determinants of long-term immune responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the concurrent impact of vaccination and emerging variants, we follow a prospective cohort of 332 patients with coronavirus disease 2019 (COVID-19) over more than a year after symptom onset. We evaluate plasma-neutralizing activity using HIV-based pseudoviruses expressing the spike of different SARS-CoV-2 variants and analyze them longitudinally using mixed-effects models. Long-term neutralizing activity is stable beyond 1 year after infection in mild/asymptomatic and hospitalized participants. However, longitudinal models suggest that hospitalized individuals generate both short- and long-lived memory B cells, while the responses of non-hospitalized individuals are dominated by long-lived B cells. In both groups, vaccination boosts responses to natural infection. Long-term (>300 days from infection) responses in unvaccinated participants show a reduced efficacy against beta, but not alpha nor delta, variants. Multivariate analysis identifies the severity of primary infection as an independent determinant of higher magnitude and lower relative cross-neutralization activity of long-term neutralizing responses. Long-term persistence (>12 months) of neutralizing antibodies against SARS-CoV-2 Severity of infection determines the magnitude and quality of neutralizing response Vaccination boosts neutralizing response to natural infection
Collapse
Affiliation(s)
- Edwards Pradenas
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, UAB, Hospital Germans Trias i Pujol, Ctra. de Canyet s/n. 2a Planta Maternal, 08916 Badalona, Catalonia, Spain
| | - Benjamin Trinité
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, UAB, Hospital Germans Trias i Pujol, Ctra. de Canyet s/n. 2a Planta Maternal, 08916 Badalona, Catalonia, Spain
| | - Víctor Urrea
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, UAB, Hospital Germans Trias i Pujol, Ctra. de Canyet s/n. 2a Planta Maternal, 08916 Badalona, Catalonia, Spain
| | - Silvia Marfil
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, UAB, Hospital Germans Trias i Pujol, Ctra. de Canyet s/n. 2a Planta Maternal, 08916 Badalona, Catalonia, Spain
| | - Ferran Tarrés-Freixas
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, UAB, Hospital Germans Trias i Pujol, Ctra. de Canyet s/n. 2a Planta Maternal, 08916 Badalona, Catalonia, Spain
| | - Raquel Ortiz
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, UAB, Hospital Germans Trias i Pujol, Ctra. de Canyet s/n. 2a Planta Maternal, 08916 Badalona, Catalonia, Spain
| | - Carla Rovirosa
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, UAB, Hospital Germans Trias i Pujol, Ctra. de Canyet s/n. 2a Planta Maternal, 08916 Badalona, Catalonia, Spain
| | - Jordi Rodon
- IRTA Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la UAB, 08193 Bellaterra, Catalonia, Spain
| | - Júlia Vergara-Alert
- IRTA Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la UAB, 08193 Bellaterra, Catalonia, Spain
| | - Joaquim Segalés
- UAB, Centre de Recerca en Sanitat Animal (IRTA-UAB), Campus de la UAB, 08193 Bellaterra, Catalonia, Spain.,Departament de Sanitat i Anatomia Animals, Facultat de Veterinària, UAB, 08193, Bellaterra, Catalonia, Spain
| | - Victor Guallar
- Barcelona Supercomputing Center, 08034 Barcelona, Catalonia, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Catalonia, Spain
| | - Alfonso Valencia
- Barcelona Supercomputing Center, 08034 Barcelona, Catalonia, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Catalonia, Spain
| | - Nuria Izquierdo-Useros
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, UAB, Hospital Germans Trias i Pujol, Ctra. de Canyet s/n. 2a Planta Maternal, 08916 Badalona, Catalonia, Spain
| | - Marc Noguera-Julian
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, UAB, Hospital Germans Trias i Pujol, Ctra. de Canyet s/n. 2a Planta Maternal, 08916 Badalona, Catalonia, Spain
| | - Jorge Carrillo
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, UAB, Hospital Germans Trias i Pujol, Ctra. de Canyet s/n. 2a Planta Maternal, 08916 Badalona, Catalonia, Spain
| | - Roger Paredes
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, UAB, Hospital Germans Trias i Pujol, Ctra. de Canyet s/n. 2a Planta Maternal, 08916 Badalona, Catalonia, Spain.,Infectious Diseases Department, Fight against AIDS Foundation (FLS), Germans Trias i Pujol Hospital, 08916 Badalona, Catalonia, Spain
| | - Lourdes Mateu
- Infectious Diseases Department, Fight against AIDS Foundation (FLS), Germans Trias i Pujol Hospital, 08916 Badalona, Catalonia, Spain
| | - Anna Chamorro
- Infectious Diseases Department, Fight against AIDS Foundation (FLS), Germans Trias i Pujol Hospital, 08916 Badalona, Catalonia, Spain
| | - Ruth Toledo
- Infectious Diseases Department, Fight against AIDS Foundation (FLS), Germans Trias i Pujol Hospital, 08916 Badalona, Catalonia, Spain
| | - Marta Massanella
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, UAB, Hospital Germans Trias i Pujol, Ctra. de Canyet s/n. 2a Planta Maternal, 08916 Badalona, Catalonia, Spain
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, UAB, Hospital Germans Trias i Pujol, Ctra. de Canyet s/n. 2a Planta Maternal, 08916 Badalona, Catalonia, Spain.,Infectious Diseases Department, Fight against AIDS Foundation (FLS), Germans Trias i Pujol Hospital, 08916 Badalona, Catalonia, Spain.,University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Catalonia, Spain
| | - Julià Blanco
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, UAB, Hospital Germans Trias i Pujol, Ctra. de Canyet s/n. 2a Planta Maternal, 08916 Badalona, Catalonia, Spain.,University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Catalonia, Spain
| |
Collapse
|
25
|
Hamady A, Lee J, Loboda ZA. Waning antibody responses in COVID-19: what can we learn from the analysis of other coronaviruses? Infection 2022; 50:11-25. [PMID: 34324165 PMCID: PMC8319587 DOI: 10.1007/s15010-021-01664-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 07/08/2021] [Indexed: 12/16/2022]
Abstract
OBJECTIVES The coronavirus disease 2019 (COVID-19), caused by the novel betacoronavirus severe acute respiratory syndrome 2 (SARS-CoV-2), was declared a pandemic in March 2020. Due to the continuing surge in incidence and mortality globally, determining whether protective, long-term immunity develops after initial infection or vaccination has become critical. METHODS/RESULTS In this narrative review, we evaluate the latest understanding of antibody-mediated immunity to SARS-CoV-2 and to other coronaviruses (SARS-CoV, Middle East respiratory syndrome coronavirus and the four endemic human coronaviruses) in order to predict the consequences of antibody waning on long-term immunity against SARS-CoV-2. We summarise their antibody dynamics, including the potential effects of cross-reactivity and antibody waning on vaccination and other public health strategies. At present, based on our comparison with other coronaviruses we estimate that natural antibody-mediated protection for SARS-CoV-2 is likely to last for 1-2 years and therefore, if vaccine-induced antibodies follow a similar course, booster doses may be required. However, other factors such as memory B- and T-cells and new viral strains will also affect the duration of both natural and vaccine-mediated immunity. CONCLUSION Overall, antibody titres required for protection are yet to be established and inaccuracies of serological methods may be affecting this. We expect that with standardisation of serological testing and studies with longer follow-up, the implications of antibody waning will become clearer.
Collapse
Affiliation(s)
- Ali Hamady
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | - JinJu Lee
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Zuzanna A Loboda
- Department of Immunology and Inflammation, Imperial College London, London, UK.
| |
Collapse
|
26
|
Zhang Y, Yang Z, Tang M, Li H, Tang T, Li G, Zhong Y, Zhang X, Wang X, Wang C. Three Specific Potential Epitopes That Could Be Recognized by T Cells of Convalescent COVID-19 Patients Were Identified From Spike Protein. Front Immunol 2022; 13:752622. [PMID: 35154095 PMCID: PMC8831549 DOI: 10.3389/fimmu.2022.752622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 01/06/2022] [Indexed: 12/17/2022] Open
Abstract
The current coronavirus disease 2019 (COVID-19) vaccines are used to prevent viral infection by inducing neutralizing antibody in the body, but according to the existing experience of severe acute respiratory syndrome coronavirus (SARS) infection, T-cell immunity could provide a longer durable protection period than antibody. The research on SARS-CoV-2-specific T-cell epitope can provide target antigen for the development and evaluation of COVID-19 vaccines, which is conducive to obtain COVID-19 vaccine that can provide long-term protection. For screening specific T-cell epitopes, a SARS-CoV-2 S protein peptide library with a peptide length of 15 amino acids was synthesized. Through flow cytometry to detect percentage of IFN-γ+ T cells after mixed COVID-19 convalescent patients' peripheral blood mononuclear cell with peptide library, seven peptides (P77, P14, P24, P38, P48, P74, and P84) that can be recognized by the T cells of COVID-19 convalescent patients were found. After excluding the nonspecific cross-reactions with unexposed population, three SARS-CoV-2-specific T-cell potential epitopes (P38, P48, and P84) were finally screened with the positive reaction rates between 15.4% and 48.0% in COVID-19 convalescent patients. This study also provided the HLA allele information of peptide-positive-response COVID-19 convalescent patients, thus predicting the population coverage of these three potential epitopes. Some HLA alleles showed higher frequency of occurrence in COVID-19 patients than in total Chinese population but no HLA alleles related to the T-cell peptide response and the severity of COVID-19. This research provides three potential T-cell epitopes that are helpful for the design and efficacy evaluation of COVID-19 vaccines. The HLA information provided by this research supplies reference significance for subsequent research such as finding the relation of HLA genotype with disease susceptibility.
Collapse
Affiliation(s)
- Yunwen Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Research and Transformation Center for Poisoning Treatment, Laboratory Science & Precision Prevention and Treatment, West China-PUMC C.C. Chen Institute of Health, Sichuan University, Chengdu, China
- AIDS Prevention and Control Department, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Zhengrong Yang
- AIDS Prevention and Control Department, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Mingyuan Tang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Research and Transformation Center for Poisoning Treatment, Laboratory Science & Precision Prevention and Treatment, West China-PUMC C.C. Chen Institute of Health, Sichuan University, Chengdu, China
| | - Hao Li
- AIDS Prevention and Control Department, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Tian Tang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Research and Transformation Center for Poisoning Treatment, Laboratory Science & Precision Prevention and Treatment, West China-PUMC C.C. Chen Institute of Health, Sichuan University, Chengdu, China
| | - Guilian Li
- AIDS Prevention and Control Department, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Yifan Zhong
- AIDS Prevention and Control Department, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Xiaomin Zhang
- AIDS Prevention and Control Department, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Xiaohui Wang
- AIDS Prevention and Control Department, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Chuan Wang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Research and Transformation Center for Poisoning Treatment, Laboratory Science & Precision Prevention and Treatment, West China-PUMC C.C. Chen Institute of Health, Sichuan University, Chengdu, China
| |
Collapse
|
27
|
Spicer KB, Glick C, Cavanaugh AM, Thoroughman D. Protective Immunity after Natural Infection with Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) - Kentucky, USA, 2020. Int J Infect Dis 2022; 114:21-28. [PMID: 34649001 PMCID: PMC8506664 DOI: 10.1016/j.ijid.2021.10.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND As vaccine supply and access remain limited in many parts of the world, understanding the duration of protection from reinfection after natural infection is important. METHODS Distinct individuals testing positive and negative for SARS-CoV-2 between March 6, 2020, and August 31, 2020, in Kentucky, USA, were identified using the Kentucky National Electronic Disease Surveillance System. Individuals were followed for occurrence of a positive test for SARS-CoV-2 from 91 days after their initial test result through December 31, 2020. Protection from reinfection provided by a prior infection was calculated and additional analyses evaluated impact of age, sex, symptom status, long-term care facility connection, testing occurrence and frequency, and time from initial infection. RESULTS The protective effect from prior infection was 80.3% (95% CI, 78.2%-82.2%) for those aged 20-59 years and 67.4% (95% CI, 62.8%-71.4%) for those aged ≥60 years. At 30-day time periods through 270 days (with limited exceptions), protection was estimated to be >75% for those aged 20-59 years and >65% for those aged ≥60 years. Factors associated with repeat positive testing included a connection to a long-term care facility, duration of potential exposure, and absence of symptoms during initial infection. CONCLUSIONS Natural infection provides substantial and persistent immunologic protection for a period of several months for most individuals, although subpopulations may be at greater risk of repeat positive testing and potential poor outcomes associated with reinfection. These subgroups include individuals aged ≥60 years, residents and staff of long-term care facilities, and those who have mild or asymptomatic illness with initial infection. Continued emphasis on vaccination and infection prevention and control strategies remains critically important in reducing the risk of reinfection and associated severe outcomes for these groups.
Collapse
Affiliation(s)
- Kevin B Spicer
- Division of Healthcare Quality Promotion, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention (CDC), USA; Kentucky Department for Public Health, USA.
| | | | - Alyson M Cavanaugh
- Kentucky Department for Public Health, USA; Epidemic Intelligence Service, CDC, USA
| | - Douglas Thoroughman
- Kentucky Department for Public Health, USA; Career Epidemiology Field Officer Program, Center for Preparedness and Response, CDC, USA
| |
Collapse
|
28
|
Bastug A, Bodur H, Aydos O, Filazi N, Oksuz E, Ozkul A. The changing dynamics of neutralizing antibody response within ten months of SARS-CoV-2 infections. J Med Virol 2021; 94:1983-1989. [PMID: 34967013 PMCID: PMC9015236 DOI: 10.1002/jmv.27544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 11/15/2021] [Accepted: 12/21/2021] [Indexed: 11/17/2022]
Abstract
There are limited data on how long neutralizing antibody (NAb) response elicited via primary SARS‐CoV‐2 infection will last. Eighty‐four serum samples were obtained from a prospective cohort of 42 laboratory‐confirmed COVID‐19 inpatients at the time of discharge from the hospital and in the late convalescent phase. A virus neutralization assay was performed to determine the presence and titers of NAbs with authentic SARS‐CoV‐2. Long‐term dynamics of NAbs and factors that may have an impact on humoral immunity were investigated. Mild and moderate/severe patients were compared. The mean sampling time was 11.12 ± 5.02 days (4–28) for the discharge test and 268.12 ± 11.65 days (247–296) for the follow‐up test. NAb response was present in 83.3% of the patients about 10 months after infection. The detectable long‐term NAb rate was significantly higher in mild patients when compared to moderate/severe patients (95.7% vs. 68.4%, p = 0.025). In the follow‐up, NAb‐positive and ‐negative patients were compared to determine the predictors of the presence of long‐term humoral immunity. The only significant factor was disease severity. Patients with mild infections have more chance to have NAbs for a longer time. Age, gender, and comorbidity did not affect long‐term NAb response. NAb titers decreased significantly over time, with an average rank of 24.0 versus 19.1 (p = 0.002). Multivariate generalized estimating equation analysis revealed that no parameter has an impact on the change of NAb titers over time. The majority of the late convalescent patients still had detectable low levels of neutralizing antibodies. The protective effect of these titers of NAbs from re‐infections needs further studies. The majority of the recovered patients (83%) had detectable NAbs up to nearly 10 months after onset. This study reveals a significant decrease in terms of NAb titers over time. Milder infections were found as the only predictor of long‐term detectable NAb response. Age, gender, and severe disease had no significant effect on changing titers of long‐term NAbs.
Collapse
Affiliation(s)
- Aliye Bastug
- Health Science University Turkey, Department of Infectious Disease and Clinical Microbiology, Ankara City Hospital, Turkey
| | - Hurrem Bodur
- Health Science University Turkey, Department of Infectious Disease and Clinical Microbiology, Ankara City Hospital, Turkey
| | - Omer Aydos
- Department of Infectious Disease and Clinical Microbiology, Ankara City Hospital, Turkey
| | - Nazlican Filazi
- Ankara University, Faculty of Veterinary Medicine, Department of Virology, Turkey
| | - Ergun Oksuz
- Department of Family Medicine, Baskent University, Ankara, Turkey
| | - Aykut Ozkul
- Ankara University, Faculty of Veterinary Medicine, Department of Virology, Turkey.,Ankara University, Biotechnology Institute, Turkey
| |
Collapse
|
29
|
Cohen JI, Burbelo PD. Reinfection With SARS-CoV-2: Implications for Vaccines. Clin Infect Dis 2021; 73:e4223-e4228. [PMID: 33338197 PMCID: PMC7799323 DOI: 10.1093/cid/ciaa1866] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/17/2020] [Indexed: 01/08/2023] Open
Abstract
Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become pandemic and the duration of protective immunity to the virus is unknown. Cases of persons reinfected with the virus are being reported with increasing frequency. At present it is unclear how common reinfection with SARS-CoV-2 is and how long serum antibodies and virus-specific T cells persist after infection. For many other respiratory virus infections, including influenza and the seasonal coronaviruses that cause colds, serum antibodies persist for only months to a few years and reinfections are very common. Here we review what is known about the duration of immunity and reinfection with coronaviruses, including SARS-CoV-2, as well as the duration of immunity to other viruses and virus vaccines. These findings have implications for the need of continued protective measures and for vaccines for persons previously infected with SARS-CoV-2.
Collapse
Affiliation(s)
- Jeffrey I Cohen
- Laboratory of Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Peter D Burbelo
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland,USA
| |
Collapse
|
30
|
Anderson RM, Vegvari C, Hollingsworth TD, Pi L, Maddren R, Ng CW, Baggaley RF. The SARS-CoV-2 pandemic: remaining uncertainties in our understanding of the epidemiology and transmission dynamics of the virus, and challenges to be overcome. Interface Focus 2021; 11:20210008. [PMID: 34956588 PMCID: PMC8504893 DOI: 10.1098/rsfs.2021.0008] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2021] [Indexed: 12/11/2022] Open
Abstract
Great progress has been made over the past 18 months in scientific understanding of the biology, epidemiology and pathogenesis of SARS-CoV-2. Extraordinary advances have been made in vaccine development and the execution of clinical trials of possible therapies. However, uncertainties remain, and this review assesses these in the context of virus transmission, epidemiology, control by social distancing measures and mass vaccination and the effect on all of these on emerging variants. We briefly review the current state of the global pandemic, focussing on what is, and what is not, well understood about the parameters that control viral transmission and make up the constituent parts of the basic reproductive number R 0. Major areas of uncertainty include factors predisposing to asymptomatic infection, the population fraction that is asymptomatic, the infectiousness of asymptomatic compared to symptomatic individuals, the contribution of viral transmission of such individuals and what variables influence this. The duration of immunity post infection and post vaccination is also currently unknown, as is the phenotypic consequences of continual viral evolution and the emergence of many viral variants not just in one location, but globally, given the high connectivity between populations in the modern world. The pattern of spread of new variants is also examined. We review what can be learnt from contact tracing, household studies and whole-genome sequencing, regarding where people acquire infection, and how households are seeded with infection since they constitute a major location for viral transmission. We conclude by discussing the challenges to attaining herd immunity, given the uncertainty in the duration of vaccine-mediated immunity, the threat of continued evolution of the virus as demonstrated by the emergence and rapid spread of the Delta variant, and the logistics of vaccine manufacturing and delivery to achieve universal coverage worldwide. Significantly more support from higher income countries (HIC) is required in low- and middle-income countries over the coming year to ensure the creation of community-wide protection by mass vaccination is a global target, not one just for HIC. Unvaccinated populations create opportunities for viral evolution since the net rate of evolution is directly proportional to the number of cases occurring per unit of time. The unit for assessing success in achieving herd immunity is not any individual country, but the world.
Collapse
Affiliation(s)
- Roy M. Anderson
- Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Carolin Vegvari
- Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - T. Déirdre Hollingsworth
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Joint Universities Pandemic and Epidemiological Research (JUNIPER) consortium, University of Leicester, Leicester, UK
| | - Li Pi
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Joint Universities Pandemic and Epidemiological Research (JUNIPER) consortium, University of Leicester, Leicester, UK
| | - Rosie Maddren
- Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Chi Wai Ng
- Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | | |
Collapse
|
31
|
Stein RA, Ometa O, Broker TR. COVID-19: The Pseudo-Environment and the Need for a Paradigm Change. Germs 2021; 11:468-477. [PMID: 35096665 DOI: 10.18683/germs.2021.1283] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Richard A Stein
- MD, PhD, NYU Tandon School of Engineering, Department of Chemical and Biomolecular Engineering, 6 MetroTech Center, Brooklyn, NY 11201, USA
| | - Oana Ometa
- PhD, Department of Journalism and Digital Media, Faculty of Political, Administrative and Communication Sciences, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Thomas R Broker
- PhD, Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama 35294-0024, USA
| |
Collapse
|
32
|
Epidemiological study in a small rural area of Veneto (Italian region) during Sars-Cov-2 Pandemia. Sci Rep 2021; 11:23247. [PMID: 34853349 PMCID: PMC8636493 DOI: 10.1038/s41598-021-02654-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 11/17/2021] [Indexed: 01/09/2023] Open
Abstract
The emergence of severe acute respiratory syndrome type 2 coronavirus (SARS-CoV-2) and its complications have demonstrated the devastating impact of a new infectious pathogen. The organisational change promulgated by the isolation of affected communities is of extreme importance to achieve effective containment of the contagion and good patient care. The epidemiological study of the population of a small rural community in the North East of Italy revealed how much the virus had circulated during Spring, 2020, and how contagion has evolved after a prolonged lockdown. In the 1st phase, NAAT (Nucleic Acid Amplification Testing) was performed in cases with more or less severe symptoms and a study was performed to trace the infection of family members. Only 0.2% of the population tested positive on NAAT, via nasopharyngeal swab during this 1st phase. In the 2nd phase a random sample of the general population were tested for circulating anti-Sars-Cov-2 immunoglobulins. This showed that approximately 97.9% of the population were negative, while 2.1% (with positive IgG at a distance) of the population had contracted the virus in a mildly symptomatic or asymptomatic form. The main symptom in subjects who developed immunity was fever. Antibodies were found in subjects with forced coexistence with quarantined or infected subjects. The mutual spatial distance by categories has shown higher relative prevalence of IgG positive and IgM negative cases in close proximity but also far from the infected, with respect to an intermediate distance. This suggests that subjects living in thinly populated areas could come in contact with the virus more likely due to intentional/relational proximity, while those living nearby could also be infected through random proximity.
Collapse
|
33
|
Gaudreault NN, Carossino M, Morozov I, Trujillo JD, Meekins DA, Madden DW, Cool K, Artiaga BL, McDowell C, Bold D, Balaraman V, Kwon T, Ma W, Henningson J, Wilson DW, Wilson WC, Balasuriya UBR, García-Sastre A, Richt JA. Experimental re-infected cats do not transmit SARS-CoV-2. Emerg Microbes Infect 2021; 10:638-650. [PMID: 33704016 PMCID: PMC8023599 DOI: 10.1080/22221751.2021.1902753] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022]
Abstract
SARS-CoV-2 is the causative agent of COVID-19 and responsible for the current global pandemic. We and others have previously demonstrated that cats are susceptible to SARS-CoV-2 infection and can efficiently transmit the virus to naïve cats. Here, we address whether cats previously exposed to SARS-CoV-2 can be re-infected with SARS-CoV-2. In two independent studies, SARS-CoV-2-infected cats were re-challenged with SARS-CoV-2 at 21 days post primary challenge (DPC) and necropsies performed at 4, 7 and 14 days post-secondary challenge (DP2C). Sentinels were co-mingled with the re-challenged cats at 1 DP2C. Clinical signs were recorded, and nasal, oropharyngeal, and rectal swabs, blood, and serum were collected and tissues examined for histologic lesions. Viral RNA was transiently shed via the nasal, oropharyngeal and rectal cavities of the re-challenged cats. Viral RNA was detected in various tissues of re-challenged cats euthanized at 4 DP2C, mainly in the upper respiratory tract and lymphoid tissues, but less frequently and at lower levels in the lower respiratory tract when compared to primary SARS-CoV-2 challenged cats at 4 DPC. Viral RNA and antigen detected in the respiratory tract of the primary SARS-CoV-2 infected cats at early DPCs were absent in the re-challenged cats. Naïve sentinels co-housed with the re-challenged cats did not shed virus or seroconvert. Together, our results indicate that cats previously infected with SARS-CoV-2 can be experimentally re-infected with SARS-CoV-2; however, the levels of virus shed was insufficient for transmission to co-housed naïve sentinels. We conclude that SARS-CoV-2 infection in cats induces immune responses that provide partial, non-sterilizing immune protection against re-infection.
Collapse
Affiliation(s)
- Natasha N. Gaudreault
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Mariano Carossino
- Louisiana Animal Disease Diagnostic Laboratory and Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Igor Morozov
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Jessie D. Trujillo
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - David A. Meekins
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Daniel W. Madden
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Konner Cool
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Bianca Libanori Artiaga
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Chester McDowell
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Dashzeveg Bold
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Velmurugan Balaraman
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Taeyong Kwon
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Wenjun Ma
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
- Department of Veterinary Pathobiology and Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA
| | - Jamie Henningson
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Dennis W. Wilson
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California-Davis, Davis, CA, USA
| | - William C. Wilson
- Arthropod Borne Animal Disease Research Unit, Agricultural Research Service, United States Department of Agriculture, Manhattan, KS, USA
| | - Udeni B. R. Balasuriya
- Louisiana Animal Disease Diagnostic Laboratory and Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Juergen A. Richt
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| |
Collapse
|
34
|
Li H, Zhang Y, Li D, Deng YQ, Xu H, Zhao C, Liu J, Wen D, Zhao J, Li Y, Wu Y, Liu S, Liu J, Hao J, Yuan F, Duo S, Qin CF, Zheng A. Enhanced protective immunity against SARS-CoV-2 elicited by a VSV vector expressing a chimeric spike protein. Signal Transduct Target Ther 2021; 6:389. [PMID: 34759261 PMCID: PMC8578532 DOI: 10.1038/s41392-021-00797-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/09/2021] [Accepted: 10/18/2021] [Indexed: 12/14/2022] Open
Abstract
SARS-CoV-2 and SARS-CoV are genetically related coronavirus and share the same cellular receptor ACE2. By replacing the VSV glycoprotein with the spikes (S) of SARS-CoV-2 and SARS-CoV, we generated two replication-competent recombinant viruses, rVSV-SARS-CoV-2 and rVSV-SARS-CoV. Using wild-type and human ACE2 (hACE2) knock-in mouse models, we found a single dose of rVSV-SARS-CoV could elicit strong humoral immune response via both intranasal (i.n.) and intramuscular (i.m.) routes. Despite the high genetic similarity between SARS-CoV-2 and SARS-CoV, no obvious cross-neutralizing activity was observed in the immunized mice sera. In macaques, neutralizing antibody (NAb) titers induced by one i.n. dose of rVSV-SARS-CoV-2 were eight-fold higher than those by a single i.m. dose. Thus, our data indicates that rVSV-SARS-CoV-2 might be suitable for i.n. administration instead of the traditional i.m. immunization in human. Because rVSV-SARS-CoV elicited significantly stronger NAb responses than rVSV-SARS-CoV-2 in a route-independent manner, we generated a chimeric antigen by replacing the receptor binding domain (RBD) of SARS-CoV S with that from the SARS-CoV-2. rVSV expressing the chimera (rVSV-SARS-CoV/2-RBD) induced significantly increased NAbs against SARS-CoV-2 in mice and macaques than rVSV-SARS-CoV-2, with a safe Th1-biased response. Serum immunized with rVSV-SARS-CoV/2-RBD showed no cross-reactivity with SARS-CoV. hACE2 mice receiving a single i.m. dose of either rVSV-SARS-CoV-2 or rVSV-SARS-CoV/2-RBD were fully protected against SARS-CoV-2 challenge without obvious lesions in the lungs. Our results suggest that transplantation of SARS-CoV-2 RBD into the S protein of SARS-CoV might be a promising antigen design for COVID-19 vaccines.
Collapse
Affiliation(s)
- Hongyue Li
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Yuhang Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Dong Li
- Shenzhen Kangtai, Biotechnology Co., Ltd, 518106, Shenzhen, Guangdong, China
| | - Yong-Qiang Deng
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Hongde Xu
- School of Pharmaceutical Sciences, Zhengzhou University, 450001, Zhengzhou, Henan, China
| | - Chaoyue Zhao
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Jiandong Liu
- Shenzhen Kangtai, Biotechnology Co., Ltd, 518106, Shenzhen, Guangdong, China
| | - Dan Wen
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Jianguo Zhao
- State Key Laboratory of Stem cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Yongchun Li
- School of Pharmaceutical Sciences, Zhengzhou University, 450001, Zhengzhou, Henan, China
| | - Yong Wu
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, 102629, Beijing, China
| | - Shujun Liu
- Laboratory Animal Center, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Jiankai Liu
- Shenzhen Kangtai, Biotechnology Co., Ltd, 518106, Shenzhen, Guangdong, China
| | - Junfeng Hao
- Core Facility for Protein Research, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China
| | - Fei Yuan
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Shuguang Duo
- Laboratory Animal Center, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China.
| | - Aihua Zheng
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, 100101, Beijing, China.
- College of Life Science, Henan Normal University, 453007, Xinxiang, China.
| |
Collapse
|
35
|
Tian X, Jiang W, Zhang H, Lu X, Li L, Liu W, Li J. Persistence of the SARS-CoV-2 Antibody Response in Asymptomatic Patients in Correctional Facilities. Front Microbiol 2021; 12:789374. [PMID: 34858383 PMCID: PMC8631518 DOI: 10.3389/fmicb.2021.789374] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 10/21/2021] [Indexed: 12/23/2022] Open
Abstract
SARS-CoV-2 has caused a global health disaster with millions of death worldwide, and the substantial proportion of asymptomatic carriers poses a huge threat to public health. The long-term antibody responses and neutralization activity during natural asymptomatic SARS-CoV-2 infection are unknown. In this study, we used enzyme-linked immunosorbent assays (ELISA) and neutralization assay with purified SARS-CoV-2S and N proteins to study the antibody responses of 156 individuals with natural asymptomatic infection. We found robust antibody responses to SARS-CoV-2 in 156 patients from 6 to 12 months. Although the antibody responses gradually decreased, S-IgG was more stable than N-IgG. S-IgG was still detected in 79% of naturally infected individuals after 12 months of infection. Moderate to potent neutralization activities were also observed in 98.74% of patients 6 months after infection. However, this proportion decreased at 8-month (46.15%) and 10-month (39.11%) after infection, respectively. Only 23.72% of patients displayed potent neutralization activity at 12 months. This study strongly supports the long-term presence of antibodies against SARS-CoV-2 in individuals with natural asymptomatic infection, although the magnitude of the antibody responses started to cripple 6 months after infection.
Collapse
Affiliation(s)
- Xiaodong Tian
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Wenguo Jiang
- Jining Center for Disease Control and Prevention, Shandong, China
| | - He Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - XiXi Lu
- Jining Center for Disease Control and Prevention, Shandong, China
| | - Libo Li
- Jining Center for Disease Control and Prevention, Shandong, China
| | - Wenjun Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- School of Life Sciences, University of Science and Technology of China, Hefei, China
- Savaid Medical School, University of the Chinese Academy of Sciences, Beijing, China
- Institute of Microbiology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Beijing, China
| | - Jing Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of the Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
36
|
Sadr S, Bafrani MA, Abdollahi A, SeyedAlinaghi SA, Mohammadnejad E, Hossienzade R, ShahmariGolestan F, Ahmadinejad Z, Salehi M, Javaherian M, Kimyaee E, Jafari F, Ghiasvand F. Distinguishing repeated polymerase chain reaction positivity from re-infections in COVID-19. Influenza Other Respir Viruses 2021; 15:742-749. [PMID: 34296828 PMCID: PMC8446995 DOI: 10.1111/irv.12883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/29/2021] [Accepted: 05/31/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Possibility of reinfection with SARS-CoV-2 changes our view on herd immunity and vaccination and can impact worldwide quarantine policies. We performed real-time polymerase chain reaction (RT-PCR) follow-up studies on recovered patients to assess possible development of reinfections and re-positivity. METHODS During a 6-month period, 202 PCR-confirmed recovering COVID-19 patients entered this study. Follow-up RT-PCR tests and symptom assessment were performed 1 month after the initial positive results. Patients who tested negative were tested again 1 and 3 months later. The serum IgG and IgM levels were measured in the last follow-up session. RESULTS In the first two follow-up sessions, 82 patients continued their participation, of which four patients tested positive. In the second follow-up 44 patients participated, three of whom tested positive. None of the patients who tested positive in the first and second follow-up session were symptomatic. In the last session, 32 patients were tested and four patients were positive, three of them were mildly symptomatic and all of them were positive for IgG. CONCLUSIONS A positive RT-PCR in a recovering patient may represent reinfection. While we did not have the resources to prove reinfection by genetic sequencing of the infective viruses, we believe presence of mild symptoms in the three patients who tested positive over 100 days after becoming asymptomatic, can be diagnosed as reinfection. The immune response developed during the first episode of infection (e.g., IgG or T-cell mediated responses that were not measured in our study) may have abated the symptoms of the reinfection, without providing complete protection.
Collapse
Affiliation(s)
- Sara Sadr
- School of MedicineMazandaran University of Medical SciencesSariIran
| | - Melika Arab Bafrani
- Students Scientific Research Center (SSRC), School of MedicineTehran University of Medical SciencesTehranIran
| | - Alireza Abdollahi
- Department of Pathology, School of Medicine, Imam Khomeini Hospital ComplexTehran University of Medical SciencesTehranIran
| | - Seyed Ahmad SeyedAlinaghi
- Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High Risk BehaviorsTehran University of Medical SciencesTehranIran
| | - Esmaeil Mohammadnejad
- Nursing Education, Department of Medical‐Surgical Nursing and Basic Science, School of Nursing and MidwiferyTehran University of Medical SciencesTehranIran
| | - Roghieh Hossienzade
- Imam Khomeini Hospital ComplexTehran University of Medical SciencesTehranIran
| | | | - Zahra Ahmadinejad
- Liver Transplantation Research Center, Department of Infectious Diseases, Imam Khomeini Hospital ComplexTehran University of Medical SciencesTehranIran
| | - Mohamadreza Salehi
- Department of Infectious Diseases, Imam Khomeini Hospital ComplexTehran University of Medical SciencesTehranIran
| | - Mohammad Javaherian
- Department of Physiotherapy, School of Rehabilitation, Liver Transplantation Research Center, Imam Khomeini Hospital ComplexTehran University of Medical SciencesTehranIran
| | - Elahe Kimyaee
- Central Laboratory, Imam Khomeini Hospital ComplexTehran University of Medical SciencesTehranIran
| | - Fatemeh Jafari
- School of MedicineIran University of Medical SciencesTehranIran
| | - Fereshteh Ghiasvand
- Liver Transplantation Research Center, Department of Infectious Diseases, Imam Khomeini Hospital ComplexTehran University of Medical SciencesTehranIran
| |
Collapse
|
37
|
Kimber C, Valk SJ, Chai KL, Piechotta V, Iannizzi C, Monsef I, Wood EM, Lamikanra AA, Roberts DJ, McQuilten Z, So-Osman C, Estcourt LJ, Skoetz N. Hyperimmune immunoglobulin for people with COVID-19. Hippokratia 2021. [DOI: 10.1002/14651858.cd015167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
| | - Sarah J Valk
- Center for Clinical Transfusion Research/Clinical Epidemiology; Sanquin/Leiden University Medical Center; Leiden Netherlands
| | - Khai Li Chai
- Transfusion Research Unit, School of Public Health and Preventive Medicine; Monash University; Melbourne Australia
| | - Vanessa Piechotta
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cochrane Haematology; Faculty of Medicine and University Hospital Cologne, University of Cologne; Cologne Germany
| | | | - Ina Monsef
- Faculty of Medicine and University Hospital Cologne, University of Cologne; Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cochrane Haematology; Cologne Germany
| | - Erica M Wood
- Transfusion Research Unit, School of Public Health and Preventive Medicine; Monash University; Melbourne Australia
| | | | - David J Roberts
- Systematic Review Initiative; NHS Blood and Transplant; Oxford UK
| | - Zoe McQuilten
- Transfusion Research Unit, School of Public Health and Preventive Medicine; Monash University; Melbourne Australia
| | - Cynthia So-Osman
- Sanquin Blood Bank; Erasmus Medical Center; Amsterdam Netherlands
| | - Lise J Estcourt
- Haematology/Transfusion Medicine; NHS Blood and Transplant; Oxford UK
| | - Nicole Skoetz
- Faculty of Medicine and University Hospital Cologne, Department of Internal Medicine; Center for Integrated Oncology, University of Cologne; Cologne Germany
| |
Collapse
|
38
|
Barnes TW, Schulte-Pelkum J, Steller L, Filchtinski D, Jenness R, Williams MR, Kober C, Manni S, Hauser T, Hahn A, Kalina U, Simon TL, Schuetz P, Roth NJ. Determination of neutralising anti-SARS-CoV-2 antibody half-life in COVID-19 convalescent donors. Clin Immunol 2021; 232:108871. [PMID: 34619377 PMCID: PMC8489294 DOI: 10.1016/j.clim.2021.108871] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 11/18/2022]
Abstract
Despite the burgeoning field of coronavirus disease-19 (COVID-19) research, the persistence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) neutralising antibodies remains unclear. This study validated two high-throughput immunological methods for use as surrogate live virus neutralisation assays and employed them to examine the half-life of SARS-CoV-2 neutralising antibodies in convalescent plasma donations made by 42 repeat donors between April and September 2020. SARS-CoV-2 neutralising antibody titres decreased over time but typically remained above the methods' diagnostic cut-offs. Using this longitudinal data, the average half-life of SARS-CoV-2 neutralising antibodies was determined to be 20.4 days. SARS-CoV-2 neutralising antibody titres appear to persist in the majority of donors for several months. Whether these titres confer protection against re-infection requires further study and is of particular relevance as COVID-19 vaccines become widely available.
Collapse
Affiliation(s)
- Thomas W Barnes
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Wankdorfstrasse 10, 3014 Bern, Switzerland
| | - Johannes Schulte-Pelkum
- Assay Design, Thermo Fisher Scientific ImmunoDiagnostics Phadia GmbH, Munzinger Strasse 7, 79111 Freiburg, Germany
| | - Laura Steller
- Assay Design, Thermo Fisher Scientific ImmunoDiagnostics Phadia GmbH, Munzinger Strasse 7, 79111 Freiburg, Germany
| | - Daniel Filchtinski
- Assay Design, Thermo Fisher Scientific ImmunoDiagnostics Phadia GmbH, Munzinger Strasse 7, 79111 Freiburg, Germany
| | - Robin Jenness
- Laboratory Operations, CSL Plasma, 1001 Corridor Park Blvd., Knoxville, TN 37932, USA
| | - Michelle R Williams
- Laboratory Operations, CSL Plasma, 1001 Corridor Park Blvd., Knoxville, TN 37932, USA
| | - Christina Kober
- Pharmaceutical Analysis and Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring Innovation GmbH, P.O. Box 1230, 35002 Marburg, Germany
| | - Sandro Manni
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Wankdorfstrasse 10, 3014 Bern, Switzerland
| | - Thomas Hauser
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Wankdorfstrasse 10, 3014 Bern, Switzerland
| | - Aaron Hahn
- Pharmaceutical Analysis and Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring Innovation GmbH, P.O. Box 1230, 35002 Marburg, Germany
| | - Uwe Kalina
- Pharmaceutical Analysis and Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring Innovation GmbH, P.O. Box 1230, 35002 Marburg, Germany
| | - Toby L Simon
- Plasma and Plasma Safety, CSL Plasma, 900 Broken Sound Parkway, Suite 400, Boca Raton, FL 33487, USA
| | - Patrick Schuetz
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Wankdorfstrasse 10, 3014 Bern, Switzerland.
| | - Nathan J Roth
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Wankdorfstrasse 10, 3014 Bern, Switzerland
| |
Collapse
|
39
|
Alwazeer D, Liu FFC, Wu XY, LeBaron TW. Combating Oxidative Stress and Inflammation in COVID-19 by Molecular Hydrogen Therapy: Mechanisms and Perspectives. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5513868. [PMID: 34646423 PMCID: PMC8505069 DOI: 10.1155/2021/5513868] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 09/20/2021] [Indexed: 02/07/2023]
Abstract
COVID-19 is a widespread global pandemic with nearly 185 million confirmed cases and about four million deaths. It is caused by an infection with the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), which primarily affects the alveolar type II pneumocytes. The infection induces pathological responses including increased inflammation, oxidative stress, and apoptosis. This situation results in impaired gas exchange, hypoxia, and other sequelae that lead to multisystem organ failure and death. As summarized in this article, many interventions and therapeutics have been proposed and investigated to combat the viral infection-induced inflammation and oxidative stress that contributes to the etiology and pathogenesis of COVID-19. However, these methods have not significantly improved treatment outcomes. This may partly be attributable to their inability at restoring redox and inflammatory homeostasis, for which molecular hydrogen (H2), an emerging novel medical gas, may complement. Herein, we systematically review the antioxidative, anti-inflammatory, and antiapoptotic mechanisms of H2. Its small molecular size and nonpolarity allow H2 to rapidly diffuse through cell membranes and penetrate cellular organelles. H2 has been demonstrated to suppress NF-κB inflammatory signaling and induce the Nrf2/Keap1 antioxidant pathway, as well as to improve mitochondrial function and enhance cellular bioenergetics. Many preclinical and clinical studies have demonstrated the beneficial effects of H2 in varying diseases, including COVID-19. However, the exact mechanisms, primary modes of action, and its true clinical effects remain to be delineated and verified. Accordingly, additional mechanistic and clinical research into this novel medical gas to combat COVID-19 complications is warranted.
Collapse
Affiliation(s)
- Duried Alwazeer
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Igdir University, 76000 Igdır, Turkey
- Research Center for Redox Applications in Foods (RCRAF), Igdir University, 76000 Igdır, Turkey
- Innovative Food Technologies Development, Application, and Research Center, Igdir University, 76000 Igdır, Turkey
| | - Franky Fuh-Ching Liu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada M5S 3M2
| | - Xiao Yu Wu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada M5S 3M2
| | - Tyler W. LeBaron
- Center of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, Bratislava, Slovakia
- Molecular Hydrogen Institute, Enoch, Utah, USA
- Department of Kinesiology and Outdoor Recreation, Southern Utah University, Cedar City, 84720 Utah, USA
| |
Collapse
|
40
|
Glennon K, Donnelly J, Knowles S, McAuliffe FM, O’Reilly A, Corcoran S, Walsh J, McMorrow R, Higgins T, Bolger L, Clinton S, O’Riordan S, Start A, Roche D, Bartels H, Malone C, McAuley K, McDermott A, Inzitari R, O’Donnell CPF, Malone F, Higgins S, De Gascun C, Doran P, Brennan DJ. Immunological assessment of SARS-CoV-2 infection in pregnancy from diagnosis to delivery: A multicentre prospective study. PLoS One 2021; 16:e0253090. [PMID: 34543278 PMCID: PMC8451988 DOI: 10.1371/journal.pone.0253090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/27/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Background Population-based data on SARS-CoV-2 infection in pregnancy and assessment of passive immunity to the neonate, is lacking. We profiled the maternal and fetal response using a combination of viral RNA from naso-pharyngeal swabs and serological assessment of antibodies against SARS-CoV-2. METHODS This multicentre prospective observational study was conducted between March 24th and August 31st 2020. Two independent cohorts were established, a symptomatic SARS-CoV-2 cohort and a cohort of asymptomatic pregnant women attending two of the largest maternity hospitals in Europe. Symptomatic women were invited to provide a serum sample to assess antibody responses. Asymptomatic pregnant women provided a nasopharyngeal swab and serum sample. RT-PCR for viral RNA was performed using the Cobas SARS-CoV-2 6800 platform (Roche). Umbilical cord bloods were obtained at delivery. Maternal and fetal serological response was measured using both the Elecsys® Anti-SARS-CoV-2 immunoassay (Roche), Abbott SARS-CoV-2 IgG Assay and the IgM Architect assay. Informed written consent was obtained from all participants. RESULTS Ten of twenty three symptomatic women had SARS-CoV-2 RNA detected on nasopharyngeal swabs. Five (5/23, 21.7%) demonstrated serological evidence of anti-SARS-CoV-2 IgG antibodies and seven (30.4%, 7/23) were positive for IgM antibodies. In the asymptomatic cohort, the prevalence of SARS-CoV-2 infection in RNA was 0.16% (1/608). IgG SARS-CoV-2 antibodies were detected in 1·67% (10/598, 95% CI 0·8%-3·1%) and IgM in 3·51% (21/598, 95% CI 2·3-5·5%). Nine women had repeat testing post the baseline test. Four (4/9, 44%) remained IgM positive and one remained IgG positive. 3 IgG anti-SARS-CoV-2 antibodies were detectable in cord bloods from babies born to five seropositive women who delivered during the study. The mean gestation at serological test was 34 weeks. The mean time between maternal serologic positivity and detection in umbilical cord samples was 28 days. CONCLUSION Using two independent serological assays, we present a comprehensive illustration of the antibody response to SARS-CoV-2 in pregnancy, and show a low prevalence of asymptomatic SARS-CoV2. Transplacental migration of anti-SARS-CoV-2 antibodies was identified in cord blood of women who demonstrated antenatal anti-SARS-CoV-2 antibodies, raising the possibility of passive immunity.
Collapse
Affiliation(s)
- Kate Glennon
- UCD School of Medicine, National Maternity Hospital, Dublin, Ireland
| | | | - Susan Knowles
- Department of Microbiology, National Maternity Hospital, Dublin, Ireland
| | - Fionnuala M. McAuliffe
- UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Ireland
| | - Alma O’Reilly
- RCSI School of Medicine, Rotunda Hospital, Dublin, Ireland
| | | | | | | | | | - Lucy Bolger
- National Maternity Hospital, Dublin, Ireland
| | | | | | - Alexander Start
- UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Ireland
| | | | | | | | - Karl McAuley
- Clinical Research Centre, UCD School of Medicine, St Vincent’s University Hospital, Dublin, Ireland
| | - Anthony McDermott
- Clinical Research Centre, UCD School of Medicine, St Vincent’s University Hospital, Dublin, Ireland
| | - Rosanna Inzitari
- Clinical Research Centre, UCD School of Medicine, St Vincent’s University Hospital, Dublin, Ireland
| | - Colm P. F. O’Donnell
- Neonatal Unit, UCD School of Medicine National Maternity Hospital, Dublin, Ireland
| | - Fergal Malone
- RCSI School of Medicine, Rotunda Hospital, Dublin, Ireland
| | - Shane Higgins
- UCD School of Medicine, National Maternity Hospital, Dublin, Ireland
- National Maternity Hospital, Dublin, Ireland
| | - Cillian De Gascun
- National Virus Reference Laboratory, University College Dublin, Dublin, Ireland
| | - Peter Doran
- Clinical Research Centre, UCD School of Medicine, St Vincent’s University Hospital, Dublin, Ireland
| | - Donal J. Brennan
- UCD School of Medicine, National Maternity Hospital, Dublin, Ireland
- Systems Biology Ireland, UCD School of Medicine, Dublin, Ireland
| |
Collapse
|
41
|
Neutralisation titres against SARS-CoV-2 are sustained 6 months after onset of symptoms in individuals with mild COVID-19. EBioMedicine 2021; 71:103519. [PMID: 34419923 PMCID: PMC8375401 DOI: 10.1016/j.ebiom.2021.103519] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/18/2021] [Accepted: 07/26/2021] [Indexed: 01/10/2023] Open
Abstract
Background Given the importance of neutralising antibodies in protection against SARS-CoV-2 infection, it is critical to assess neutralisation persistence long-term following recovery. This study investigated neutralisation titres against SARS-CoV-2 up to 6 months post-symptom onset in individuals with mild COVID-19. Methods Plasma neutralisation titres in convalescent COVID-19 individuals were determined at baseline and 6 months post-symptom onset using a cell culture infectious SARS-CoV-2 assay. Total SARS-CoV-2 spike-specific IgG and IgA binding was measured using a lectin capture ELISA and compared between timepoints and correlated to neutralising titres. Findings All 48 convalescent COVID-19 individuals were found to have detectable SARS-CoV-2 50% inhibitory dilution neutralisation titres (ID50) at baseline and 6 months post-symptom onset with mean ID50 of 1/943 and 1/411, respectively. SARS-CoV-2 neutralisation titres peaked within 1-2 months post-symptom onset. However, 50% of individuals showed comparable ID50 at baseline and 6 months post-symptom onset. Both SARS-CoV-2 spike-specific IgG and IgA levels correlated well with neutralising titres. IgG binding was found to be sustained up to 6 months post-symptom onset, whereas IgA levels declined. Interpretation This study demonstrates durability of SARS-CoV-2 spike-specific IgG and neutralisation responses following recovery from mild COVID-19. Thus, all subjects included in this study might potentially have protective levels of neutralising antibodies 6 months post-symptom onset. This study also demonstrates a relationship between spike-specific IgA and neutralisation decline, with implications for long-term protection against SARS-CoV-2 infection. Funding Novo Nordisk Foundation, Independent Research Fund Denmark and Danish Agency for Science and Higher Education.
Collapse
|
42
|
Pan Y, Jiang X, Yang L, Chen L, Zeng X, Liu G, Tang Y, Qian C, Wang X, Cheng F, Lin J, Wang X, Li Y. SARS-CoV-2-specific immune response in COVID-19 convalescent individuals. Signal Transduct Target Ther 2021; 6:256. [PMID: 34234102 PMCID: PMC8261819 DOI: 10.1038/s41392-021-00686-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 06/10/2021] [Accepted: 06/20/2021] [Indexed: 02/07/2023] Open
Abstract
We collected blood from coronavirus disease 2019 (COVID-19) convalescent individuals and investigated SARS-CoV-2-specific humoral and cellular immunity in these discharged patients. Follow-up analysis in a cohort of 171 patients at 4-11 months after the onset revealed high levels of IgG antibodies. A total of 78.1% (164/210) of the specimens tested positive for neutralizing antibody (NAb). SARS-CoV-2 antigen peptide pools-stimulated-IL-2 and -IFN-γ response can distinguish COVID-19 convalescent individuals from healthy donors. Interestingly, NAb survival was significantly affected by the antigen peptide pools-stimulated-IL-2 response, -IL-8 response, and -IFN-γ response. The antigen peptide pools-activated CD8+ T cell counts were correlated with NAb. The antigen peptide pools-activated natural killer (NK) cell counts in convalescent individuals were correlated with NAb and disease severity. Our data suggested that the development of NAb is associated with the activation of T cells and NK cells. Our work provides a basis for further analysis of the protective immunity to SARS-CoV-2 and for understanding the pathogenesis of COVID-19. It also has implications for the development of an effective vaccine for SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Yunbao Pan
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China
| | - Xianghu Jiang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China
| | - Liu Yang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
| | - Liangjun Chen
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
| | - Xiaojiao Zeng
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
| | - Guohong Liu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yueting Tang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
| | - Chungen Qian
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xinming Wang
- Reagent R&D Center, Autobio Diagnostics Co., Ltd, Zhengzhou, Henan, China
| | - Fangming Cheng
- Reagent R&D Center, Shenzhen YHLO Biotech Co., Ltd, Shenzhen, Guangdong, China
| | - Jun Lin
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China.
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China.
| | - Xinghuan Wang
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China.
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China.
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China.
| | - Yirong Li
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China.
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
43
|
Grandjean L, Saso A, Ortiz AT, Lam T, Hatcher J, Thistlethwayte R, Harris M, Best T, Johnson M, Wagstaffe H, Ralph E, Mai A, Colijn C, Breuer J, Buckland M, Gilmour K, Goldblatt D. Long-Term Persistence of Spike Antibody and Predictive Modeling of Antibody Dynamics Following Infection with SARS-CoV-2. Clin Infect Dis 2021; 74:1220-1229. [PMID: 34218284 PMCID: PMC8994590 DOI: 10.1093/cid/ciab607] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Indexed: 01/08/2023] Open
Abstract
Background Antibodies to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been shown to neutralize the virus in vitro and prevent disease in animal challenge models on reexposure. However, the current understanding of SARS-CoV-2 humoral dynamics and longevity is conflicting. Methods The COVID-19 Staff Testing of Antibody Responses Study (Co-Stars) prospectively enrolled 3679 healthcare workers to comprehensively characterize the kinetics of SARS-CoV-2 spike protein (S), receptor-binding domain, and nucleoprotein (N) antibodies in parallel. Participants screening seropositive had serial monthly serological testing for a maximum of 7 months with the Meso Scale Discovery Assay. Survival analysis determined the proportion of seroreversion, while 2 hierarchical gamma models predicted the upper and lower bounds of long-term antibody trajectory. Results A total of 1163 monthly samples were provided from 349 seropositive participants. At 200 days after symptoms, >95% of participants had detectable S antibodies, compared with 75% with detectable N antibodies. S antibody was predicted to remain detectable in 95% of participants until 465 days (95% confidence interval, 370–575 days) using a “continuous-decay” model and indefinitely using a “decay-to-plateau” model to account for antibody secretion by long-lived plasma cells. S-antibody titers were correlated strongly with surrogate neutralization in vitro (R2 = 0.72). N antibodies, however, decayed rapidly with a half-life of 60 days (95% confidence interval, 52–68 days). Conclusions The Co-Stars data presented here provide evidence for long-term persistence of neutralizing S antibodies. This has important implications for the duration of functional immunity after SARS-CoV-2 infection. In contrast, the rapid decay of N antibodies must be considered in future seroprevalence studies and public health decision-making. This is the first study to establish a mathematical framework capable of predicting long-term humoral dynamics after SARS-CoV-2 infection. Clinical Trials Registration NCT04380896.
Collapse
Affiliation(s)
- Louis Grandjean
- Department of Infection, Inflammation and Immunity, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, UK.,Department of Infectious Diseases, Great Ormond Street Hospital, Great Ormond Street, London
| | - Anja Saso
- Department of Infectious Diseases, Great Ormond Street Hospital, Great Ormond Street, London.,Department of Tropical and Infectious diseases; LSHTM, Keppel St, Bloomsbury, London.,MRC Gambia at LSHTM, PO Box 273, Fajara, The Gambia
| | - Arturo Torres Ortiz
- Department of Infection, Inflammation and Immunity, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, UK.,Department of Medicine, Imperial College, Paddington, London
| | - Tanya Lam
- Department of Infectious Diseases, Great Ormond Street Hospital, Great Ormond Street, London
| | - James Hatcher
- Department of Microbiology, Great Ormond Street Hospital, Great Ormond Street, London
| | | | - Mark Harris
- Quality Improvement, Great Ormond Street Hospital, Great Ormond Street, London
| | - Timothy Best
- Department of Medicine, Imperial College, Paddington, London
| | - Marina Johnson
- Department of Infection, Inflammation and Immunity, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, UK
| | - Helen Wagstaffe
- Department of Infection, Inflammation and Immunity, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, UK
| | - Elizabeth Ralph
- Clinical Immunology, Camelia Botnar Laboratories, Great Ormond Street Hospital, Great Ormond Street, London
| | - Annabelle Mai
- Clinical Immunology, Camelia Botnar Laboratories, Great Ormond Street Hospital, Great Ormond Street, London
| | - Caroline Colijn
- Department of Mathematics, Simon Fraser University, Vancouver, British Colombia, Canada
| | - Judith Breuer
- Department of Infection, Inflammation and Immunity, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, UK
| | - Matthew Buckland
- Clinical Immunology, Camelia Botnar Laboratories, Great Ormond Street Hospital, Great Ormond Street, London
| | - Kimberly Gilmour
- Clinical Immunology, Camelia Botnar Laboratories, Great Ormond Street Hospital, Great Ormond Street, London
| | - David Goldblatt
- Department of Infection, Inflammation and Immunity, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, UK
| | | |
Collapse
|
44
|
Retrospective of International Serological Studies on the Formation and Dynamics of the Humoral Immune Response to SARS-CoV-2: from 2020 to 2021. ACTA BIOMEDICA SCIENTIFICA 2021. [DOI: 10.29413/abs.2021-6.2.5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Last year the COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has started. The new coronavirus is highly contagious and causes severe complications. The mechanisms of humoral immunity and kinetics of SARS-CoV-2 specific antibodies in a population are not well understood. Therefore, we aimed to summarize and analyze numerous global and Russian serological studies for understanding dynamics of the SARSCoV-2 humoral immune response and getting an accurate picture of the seroprevalence to SARS-CoV-2 in the world population. The PubMed and e-library databases were searched from February 2020 to March 2021 using terms “SARSCoV-2”, “antibodies”, “humoral immunity”. At the beginning of the pandemic first studies were cross-sectional by design and were responsible for determination of the seropositivity and for understanding the fundamental humoral immunity parameters of SARS-CoV-2. Since then, longitudinal seroepidemiological studies have been studying antibody kinetics. Seroconversion time for IgM, IgG antibodies varies, but most researchers report the seroconversion of IgM from the 1st to 14th days after the onset of clinical manifestations, and the seroconversion for IgG is around the 14th day with a concentration peak by the 21st day. Regarding seroprevalence we may say about low herd immunity at the COVID-19 pandemic. Thus, global seroprevalence is about 10 %, and more than 20 % for regions with high incidence and among healthcare workers. Seroprevalence studies have to be continued for more accurate monitoring of long-term humoral immunity to SARS-CoV-2, because the majority of the world’s population is still susceptible to SARS-CoV-2 infection.
Collapse
|
45
|
Jamiruddin MR, Haq MA, Tomizawa K, Kobatake E, Mie M, Ahmed S, Khandker SS, Ali T, Jahan N, Oishee MJ, Khondoker MU, Sil BK, Haque M, Adnan N. Longitudinal Antibody Dynamics Against Structural Proteins of SARS-CoV-2 in Three COVID-19 Patients Shows Concurrent Development of IgA, IgM, and IgG. J Inflamm Res 2021; 14:2497-2506. [PMID: 34163208 PMCID: PMC8214341 DOI: 10.2147/jir.s313188] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/19/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Dynamics and persistence of neutralizing and non-neutralizing antibodies can give us the knowledge required for serodiagnosis, disease management, and successful vaccine design and development. The disappearance of antibodies, absence of humoral immunity activation, and sporadic reinfection cases emphasize the importance of longitudinal antibody dynamics against variable structural antigens. METHODS In this study, twenty-five healthy subjects working in a SARS-COV-2 serodiagnostic assay development project were enrolled, and their sign and symptoms were followed up to six months. Three subjects showed COVID-19-like symptoms, and three subjects' antibody dynamics were followed over 120 days by analyzing 516 samples. We have developed 12 different types of in-house ELISAs to observe the kinetics of IgG, IgM, and IgA against four SARS-CoV-2 proteins, namely nucleocapsid, RBD, S1, and whole spike (S1+S2). For the development of these assays, 30-104 pre-pandemic samples were taken as negative controls and 83 RT-qPCR positive samples as positive ones. RESULTS All three subjects presented COVID-19-like symptoms twice, with mild symptoms in the first episode were severe in the second, and RT-qPCR confirmed the latter. The initial episode did not culminate with any significant antibody development, while a multifold increase in IgG antibodies characterized the second episode. Interestingly, IgG antibody development concurrent with IgM and IgA and persisted, whereas the latter two weans off rather quickly if appeared. CONCLUSION Antibody kinetics observed in this study can provide a pathway to the successful development of sero-diagnostics and epidemiologists to predict the fate of vaccination currently in place.
Collapse
Affiliation(s)
| | - Md Ahsanul Haq
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhaka, 1205, Bangladesh
| | - Kazuhito Tomizawa
- Department of Molecular Physiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-0811, Japan
| | - Eiry Kobatake
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, 226-8502, Japan
| | - Masayasu Mie
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, 226-8502, Japan
| | - Sohel Ahmed
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh
| | - Shahad Saif Khandker
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhaka, 1205, Bangladesh
| | - Tamanna Ali
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhaka, 1205, Bangladesh
| | - Nowshin Jahan
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhaka, 1205, Bangladesh
| | | | | | - Bijon Kumar Sil
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhaka, 1205, Bangladesh
| | - Mainul Haque
- The Unit of Pharmacology, Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia (National Defence University of Malaysia), Kuala Lumpur, 57000, Malaysia
| | - Nihad Adnan
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh
| |
Collapse
|
46
|
Izak M, Gendelman V, Bransburg-Zabary S, Stoyanov E, Gat R, Cohen D, Chen J, Maor Y, Benov A, Lev B, Zimhony O, Shinar E. Qualifying coronavirus disease 2019 convalescent plasma donors in Israel. Vox Sang 2021; 117:185-192. [PMID: 34125976 PMCID: PMC8447161 DOI: 10.1111/vox.13162] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/18/2021] [Accepted: 05/18/2021] [Indexed: 12/19/2022]
Abstract
Background and Objectives Passive immunization using investigational COVID‐19 convalescent plasma (CCP) is a promising therapeutic strategy and could improve outcome if transfused early and contain high levels of anti‐SARS‐CoV‐2 antibodies. We report the management of a national CCP collection and distribution program in Israel. Materials and Methods From 1 April 2020 to 15 January 2021, 4020 volunteer donors donated 5221 CCP units and 837 (20.8%) donors donated more than once. Anti‐nucleocapsid IgG antibodies were determined using chemiluminescent immunoassay method (Abbott). A statistical model based on repeated IgG tests in sequential donations was created to predict the time of antibody decline below sample/cut‐off (S/CO) level of 4.0. Results Ninety‐six percent of CCP donors suffered a mild disease or were asymptomatic. Older donors had higher antibody levels. Higher antibody levels (S/CO ≥4) were detected in 35.2% of the donors. Low positive (S/CO ≥1.4–3.99) were found in 37%, and 27.8% had undetectable antibodies (S/CO ≤1.4). The model predicted decrease antibody thresholds of 0.55%/day since the first CCP donation, providing guidance for the effective timing of future collections from donors with high antibody levels. Conclusions An efficient CCP collection and distribution program was achieved, based on performing initial and repeated plasma collections, preferably from donors with higher antibody levels, and only antibody‐rich units were supplied for therapeutic use. The inventory met the quantity and quality standards of the authorities, enabled to respond to the growing demand of the medical system and provide a product that may contribute to improve prognosis in patients with COVID‐19.
Collapse
Affiliation(s)
- Marina Izak
- National Blood Services, Magen David Adom, Ramat Gan, Israel
| | | | | | | | - Roni Gat
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Clinical Research Center, Soroka University Medical Center, Beer Sheva, Israel
| | - Daniel Cohen
- School of Public Health, Tel Aviv University, Tel Aviv, Israel
| | - Jacob Chen
- Trauma and Combat Medicine Branch, Israel Defense Forces Medical Corps, Ramat Gan, Israel.,Hospital Management, Meir Medical Center, KfarSaba, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yasmin Maor
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Infectious Disease Unit, Wolfson Medical Center, Holon, Israel
| | - Avi Benov
- Trauma and Combat Medicine Branch, Israel Defense Forces Medical Corps, Ramat Gan, Israel.,The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Boaz Lev
- Epidemic Unit, Ministry of Health, Jerusalem, Israel
| | - Oren Zimhony
- Infectious Disease Unit, Kaplan Medical Center, Rehovot, Israel
| | - Eilat Shinar
- National Blood Services, Magen David Adom, Ramat Gan, Israel.,Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
47
|
Chaudhury S, Hutter J, Bolton JS, Hakre S, Mose E, Wooten A, O’Connell W, Hudak J, Krebs SJ, Darden JM, Regules JA, Murray CK, Modjarrad K, Scott P, Peel S, Bergmann-Leitner ES. Serological profiles of pan-coronavirus-specific responses in COVID-19 patients using a multiplexed electro-chemiluminescence-based testing platform. PLoS One 2021; 16:e0252628. [PMID: 34081747 PMCID: PMC8174743 DOI: 10.1371/journal.pone.0252628] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 05/19/2021] [Indexed: 01/15/2023] Open
Abstract
Serological assessment of SARS-CoV-2 specific responses are an essential tool for determining the prevalence of past SARS-CoV-2 infections in the population especially when testing occurs after symptoms have developed and limited contact tracing is in place. The goal of our study was to test a new 10-plex electro-chemiluminescence-based assay to measure IgM and IgG responses to the spike proteins from multiple human coronaviruses including SARS-CoV-2, assess the epitope specificity of the SARS-CoV-2 antibody response against full-length spike protein, receptor-binding domain and N-terminal domain of the spike protein, and the nucleocapsid protein. We carried out the assay on samples collected from three sample groups: subjects diagnosed with COVID-19 from the U.S. Army hospital at Camp Humphreys in Pyeongtaek, South Korea; healthcare administrators from the same hospital but with no reported diagnosis of COVID-19; and pre-pandemic samples. We found that the new CoV-specific multiplex assay was highly sensitive allowing plasma samples to be diluted 1:30,000 with a robust signal. The reactivity of IgG responses to SARS-CoV-2 nucleocapsid protein and IgM responses to SARS-CoV-2 spike protein could distinguish COVID-19 samples from non-COVID-19 and pre-pandemic samples. The data from the three sample groups also revealed a unique pattern of cross-reactivity between SARS-CoV-2 and SARS-CoV-1, MERS-CoV, and seasonal coronaviruses HKU1 and OC43. Our findings show that the CoV-2 IgM response is highly specific while the CoV-2 IgG response is more cross-reactive across a range of human CoVs and also showed that IgM and IgG responses show distinct patterns of epitope specificity. In summary, this multiplex assay was able to distinguish samples by COVID-19 status and characterize distinct trends in terms of cross-reactivity and fine-specificity in antibody responses, underscoring its potential value in diagnostic or serosurveillance efforts.
Collapse
Affiliation(s)
- Sidhartha Chaudhury
- Center Enabling Capabilities, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Jack Hutter
- Clinical Trials Center, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Jessica S. Bolton
- Immunology Core, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for The Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Shilpa Hakre
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Evelyn Mose
- Brian D. Allgood Army Community Hospital, Camp Humphreys, Pyeongtaek, South Korea
| | - Amy Wooten
- Brian D. Allgood Army Community Hospital, Camp Humphreys, Pyeongtaek, South Korea
| | - William O’Connell
- Brian D. Allgood Army Community Hospital, Camp Humphreys, Pyeongtaek, South Korea
| | - Joseph Hudak
- Brian D. Allgood Army Community Hospital, Camp Humphreys, Pyeongtaek, South Korea
| | - Shelly J. Krebs
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Janice M. Darden
- Henry M. Jackson Foundation for The Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Jason A. Regules
- Immunology Core, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | | | - Kayvon Modjarrad
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Paul Scott
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Sheila Peel
- Diagnostics and Countermeasures Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Elke S. Bergmann-Leitner
- Immunology Core, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| |
Collapse
|
48
|
Alfego D, Sullivan A, Poirier B, Williams J, Adcock D, Letovsky S. A population-based analysis of the longevity of SARS-CoV-2 antibody seropositivity in the United States. EClinicalMedicine 2021; 36:100902. [PMID: 34056568 PMCID: PMC8143650 DOI: 10.1016/j.eclinm.2021.100902] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/21/2021] [Accepted: 04/27/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND This cross-sectional study aimed to track population-based SARS-CoV-2 antibody seropositivity duration across the United States using observational data from a national clinical laboratory registry of patients tested by nucleic acid amplification (NAAT) and serologic assays. Knowledge of antibody seropositivity and its duration may help dictate post-pandemic planning. METHODS Using assays to detect antibodies to either nucleocapsid (N) or spike (S) proteins performed on specimens from 39,086 individuals with confirmed positive COVID-19 by reverse transcription-polymerase chain reaction (RT-PCR) from March 2020 to January 2021, we analyzed nationwide seropositivity rates of IgG up to 300 days following patients' initial positive NAAT test. Linear regression identified trends in seropositivity rates and logistic regression tested positive predictability by age, sex, assay type and days post-infection. FINDINGS Seropositivity of IgG antibodies to both SARS-CoV-2 S and N-proteins followed a linear trend reaching approximately 90% positivity at 21 days post-index. The rate of N-protein seropositivity declined at a sharper rate, decaying to 68·2% [95% CI: 63·1-70·8%] after 293 days, while S-antibody seropositivity maintained a rate of 87·8% [95% CI: 86·3-89·1%] through 300 days. In addition to antigen type and the number of days post-positive PCR, age and gender were also significant factors in seropositivity prediction, with those under 65 years of age showing a more sustained seropositivity rate. INTERPRETATION Observational data from a national clinical laboratory, though limited by an epidemiological view of the U.S. population, offer an encouraging timeline for the development and sustainability of antibodies up to ten months from natural infection and could inform post-pandemic planning.
Collapse
Affiliation(s)
- David Alfego
- Laboratory Corporation of America® Holdings, 3595 Johns Hopkins Court, San Diego, CA 92121, United States
| | - Adam Sullivan
- Laboratory Corporation of America® Holdings, Burlington, NC, United States
| | - Brian Poirier
- Laboratory Corporation of America® Holdings, Burlington, NC, United States
| | - Jonathan Williams
- Laboratory Corporation of America® Holdings, Burlington, NC, United States
| | - Dorothy Adcock
- Laboratory Corporation of America® Holdings, Burlington, NC, United States
| | - Stanley Letovsky
- Laboratory Corporation of America® Holdings, Burlington, NC, United States
| |
Collapse
|
49
|
Ekelund O, Ekblom K, Somajo S, Pattison-Granberg J, Olsson K, Petersson A. High-throughput immunoassays for SARS-CoV-2 - considerable differences in performance when comparing three methods. Infect Dis (Lond) 2021; 53:805-810. [PMID: 34053400 PMCID: PMC8171012 DOI: 10.1080/23744235.2021.1931434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background The recently launched high-throughput assays for detecting antibodies against SARS-CoV-2 has contributed to the managing strategies for the COVID-19 pandemic. This study aimed to investigate the performance of three high-throughput assays and one rapid lateral flow test relative to regulatory authorities' recommended criteria. Methods A total of 315 samples, including 150 pre-pandemic samples, 152 samples from SARS-CoV-2 RT-PCR positive individuals and 13 potentially cross-reactive samples were analysed with SARS-CoV-2 IgG (Abbott, Abbott Park, IL), Elecsys Anti-SARS-CoV-2 (Roche, Solna, Sweden), LIAISON SARS-CoV-2 S1/S2 IgG (DiaSorin, Saluggia, Italy) and 2019-nCOV IgG/IgM Rapid Test (Dynamiker Biotechnology Co., Tianjin, China). Results All assays performed with a high level of specificity ranging from 96.7% to 99.3%. Sensitivity differed more between the assays, Roche exhibiting the highest sensitivity of 98.7%. The corresponding figures for Abbott, DiaSorin and Dynamiker Biotechnology were 80.9%, 89.0% and 72.4%, respectively. Conclusions The results of the evaluated SARS-CoV-2 assays vary considerably, as well as their ability to fulfil the performance criteria proposed by regulatory authorities. Introduction into clinical use in low-prevalent settings, should, therefore, be made with caution.
Collapse
Affiliation(s)
- Oskar Ekelund
- Department of Clinical Microbiology, Växjö Central Hospital, Växjö, Sweden.,Department of Clinical Microbiology, Blekinge County Hospital, Karlskrona, Sweden
| | - Kim Ekblom
- Department of Clinical Chemistry and Transfusion Medicine, Växjö Central Hospital, Växjö, Sweden.,Department of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, Sweden
| | - Sofia Somajo
- Department of Clinical Microbiology, Blekinge County Hospital, Karlskrona, Sweden
| | | | - Karl Olsson
- Department of Clinical Microbiology, Blekinge County Hospital, Karlskrona, Sweden
| | - Annika Petersson
- Department of Clinical Chemistry and Transfusion Medicine, Växjö Central Hospital, Växjö, Sweden
| |
Collapse
|
50
|
Piechotta V, Iannizzi C, Chai KL, Valk SJ, Kimber C, Dorando E, Monsef I, Wood EM, Lamikanra AA, Roberts DJ, McQuilten Z, So-Osman C, Estcourt LJ, Skoetz N. Convalescent plasma or hyperimmune immunoglobulin for people with COVID-19: a living systematic review. Cochrane Database Syst Rev 2021; 5:CD013600. [PMID: 34013969 PMCID: PMC8135693 DOI: 10.1002/14651858.cd013600.pub4] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Convalescent plasma and hyperimmune immunoglobulin may reduce mortality in patients with viral respiratory diseases, and are being investigated as potential therapies for coronavirus disease 2019 (COVID-19). A thorough understanding of the current body of evidence regarding benefits and risks of these interventions is required. OBJECTIVES: Using a living systematic review approach, to assess whether convalescent plasma or hyperimmune immunoglobulin transfusion is effective and safe in the treatment of people with COVID-19; and to maintain the currency of the evidence. SEARCH METHODS To identify completed and ongoing studies, we searched the World Health Organization (WHO) COVID-19 Global literature on coronavirus disease Research Database, MEDLINE, Embase, the Cochrane COVID-19 Study Register, the Epistemonikos COVID-19 L*OVE Platform, and trial registries. Searches were done on 17 March 2021. SELECTION CRITERIA We included randomised controlled trials (RCTs) evaluating convalescent plasma or hyperimmune immunoglobulin for COVID-19, irrespective of disease severity, age, gender or ethnicity. For safety assessments, we also included non-controlled non-randomised studies of interventions (NRSIs) if 500 or more participants were included. We excluded studies that included populations with other coronavirus diseases (severe acute respiratory syndrome (SARS) or Middle East respiratory syndrome (MERS)), as well as studies evaluating standard immunoglobulin. DATA COLLECTION AND ANALYSIS We followed standard Cochrane methodology. To assess bias in included studies, we used the Cochrane 'Risk of Bias 2' tool for RCTs, and for NRSIs, the assessment criteria for observational studies, provided by Cochrane Childhood Cancer. We rated the certainty of evidence, using the GRADE approach, for the following outcomes: all-cause mortality, improvement and worsening of clinical status (for individuals with moderate to severe disease), development of severe clinical COVID-19 symptoms (for individuals with asymptomatic or mild disease), quality of life (including fatigue and functional independence), grade 3 or 4 adverse events, and serious adverse events. MAIN RESULTS We included 13 studies (12 RCTs, 1 NRSI) with 48,509 participants, of whom 41,880 received convalescent plasma. We did not identify any completed studies evaluating hyperimmune immunoglobulin. We identified a further 100 ongoing studies evaluating convalescent plasma or hyperimmune immunoglobulin, and 33 studies reporting as being completed or terminated. Individuals with a confirmed diagnosis of COVID-19 and moderate to severe disease Eleven RCTs and one NRSI investigated the use of convalescent plasma for 48,349 participants with moderate to severe disease. Nine RCTs compared convalescent plasma to placebo treatment or standard care alone, and two compared convalescent plasma to standard plasma (results not included in abstract). Effectiveness of convalescent plasma We included data on nine RCTs (12,875 participants) to assess the effectiveness of convalescent plasma compared to placebo or standard care alone. Convalescent plasma does not reduce all-cause mortality at up to day 28 (risk ratio (RR) 0.98, 95% confidence interval (CI) 0.92 to 1.05; 7 RCTs, 12,646 participants; high-certainty evidence). It has little to no impact on clinical improvement for all participants when assessed by liberation from respiratory support (RR not estimable; 8 RCTs, 12,682 participants; high-certainty evidence). It has little to no impact on the chance of being weaned or liberated from invasive mechanical ventilation for the subgroup of participants requiring invasive mechanical ventilation at baseline (RR 1.04, 95% CI 0.57 to 1.93; 2 RCTs, 630 participants; low-certainty evidence). It does not reduce the need for invasive mechanical ventilation (RR 0.98, 95% CI 0.89 to 1.08; 4 RCTs, 11,765 participants; high-certainty evidence). We did not identify any subgroup differences. We did not identify any studies reporting quality of life, and therefore, do not know whether convalescent plasma has any impact on quality of life. One RCT assessed resolution of fatigue on day 7, but we are very uncertain about the effect (RR 1.21, 95% CI 1.02 to 1.42; 309 participants; very low-certainty evidence). Safety of convalescent plasma We included results from eight RCTs, and one NRSI, to assess the safety of convalescent plasma. Some of the RCTs reported on safety data only for the convalescent plasma group. We are uncertain whether convalescent plasma increases or reduces the risk of grade 3 and 4 adverse events (RR 0.90, 95% CI 0.58 to 1.41; 4 RCTs, 905 participants; low-certainty evidence), and serious adverse events (RR 1.24, 95% CI 0.81 to 1.90; 2 RCTs, 414 participants; low-certainty evidence). A summary of reported events of the NRSI (reporting safety data for 20,000 of 35,322 transfused participants), and four RCTs reporting safety data only for transfused participants (6125 participants) are included in the full text. Individuals with a confirmed diagnosis of SARS-CoV-2 infection and asymptomatic or mild disease We identified one RCT reporting on 160 participants, comparing convalescent plasma to placebo treatment (saline). Effectiveness of convalescent plasma We are very uncertain about the effect of convalescent plasma on all-cause mortality (RR 0.50, 95% CI 0.09 to 2.65; very low-certainty evidence). We are uncertain about the effect of convalescent plasma on developing severe clinical COVID-19 symptoms (RR not estimable; low-certainty evidence). We identified no study reporting quality of life. Safety of convalescent plasma We do not know whether convalescent plasma is associated with a higher risk of grade 3 or 4 adverse events (very low-certainty evidence), or serious adverse events (very low-certainty evidence). This is a living systematic review. We search weekly for new evidence and update the review when we identify relevant new evidence. Please refer to the Cochrane Database of Systematic Reviews for the current status of this review. AUTHORS' CONCLUSIONS We have high certainty in the evidence that convalescent plasma for the treatment of individuals with moderate to severe disease does not reduce mortality and has little to no impact on measures of clinical improvement. We are uncertain about the adverse effects of convalescent plasma. While major efforts to conduct research on COVID-19 are being made, heterogeneous reporting of outcomes is still problematic. There are 100 ongoing studies and 33 studies reporting in a study registry as being completed or terminated. Publication of ongoing studies might resolve some of the uncertainties around hyperimmune immunoglobulin therapy for people with any disease severity, and convalescent plasma therapy for people with asymptomatic or mild disease.
Collapse
Affiliation(s)
- Vanessa Piechotta
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Claire Iannizzi
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Khai Li Chai
- Transfusion Research Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Sarah J Valk
- Jon J van Rood Center for Clinical Transfusion Research, Sanquin/Leiden University Medical Center, Leiden, Netherlands
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Catherine Kimber
- Systematic Review Initiative, NHS Blood and Transplant, Oxford, UK
| | - Elena Dorando
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ina Monsef
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Erica M Wood
- Transfusion Research Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | | | - David J Roberts
- Systematic Review Initiative, NHS Blood and Transplant, Oxford, UK
| | - Zoe McQuilten
- Transfusion Research Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Cynthia So-Osman
- Sanquin Blood Bank, Amsterdam, Netherlands
- Erasmus Medical Centre, Rotterdam, Netherlands
| | - Lise J Estcourt
- Haematology/Transfusion Medicine, NHS Blood and Transplant, Oxford, UK
| | - Nicole Skoetz
- Cochrane Cancer, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|