1
|
Movsisyan M, Truzyan N, Kasparova I, Chopikyan A, Sawaqed R, Bedross A, Sukiasyan M, Dilbaryan K, Shariff S, Kantawala B, Hakobjanyan G, Petrosyan G, Hakobyan A, Yenkoyan K. Tracking the evolution of anti-SARS-CoV-2 antibodies and long-term humoral immunity within 2 years after COVID-19 infection. Sci Rep 2024; 14:13417. [PMID: 38862731 PMCID: PMC11167004 DOI: 10.1038/s41598-024-64414-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 06/08/2024] [Indexed: 06/13/2024] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that gave rise to COVID-19 infection produced a worldwide health crisis. The virus can cause a serious or even fatal disease. Comprehending the complex immunological responses triggered by SARS-CoV-2 infection is essential for identifying pivotal elements that shape the course of the disease and its enduring effects on immunity. The span and potency of antibody responses provide valuable perspicuity into the resilience of post-infection immunity. The analysis of existing literature reveals a diverse controversy, confining varying data about the persistence of particular antibodies as well as the multifaceted factors that impact their development and titer, Within this study we aimed to understand the dynamics of anti-SARS-CoV-2 antibodies against nucleocapsid (anti-SARS-CoV-2 (N)) and spike (anti-SARS-CoV-2 (N)) proteins in long-term immunity in convalescent patients, as well as the factors influencing the production and kinetics of those antibodies. We collected 6115 serum samples from 1611 convalescent patients at different post-infection intervals up to 21 months Study showed that in the fourth month, the anti-SARS-CoV-2 (N) exhibited their peak mean value, demonstrating a 79% increase compared to the initial month. Over the subsequent eight months, the peak value experienced a modest decline, maintaining a relatively elevated level by the end of study. Conversely, anti-SARS-CoV-2 (S) exhibited a consistent increase at each three-month interval over the 15-month period, culminating in a statistically significant peak mean value at the study's conclusion. Our findings demonstrate evidence of sustained seropositivity rates for both anti-SARS-CoV-2 (N) and (S), as well as distinct dynamics in the long-term antibody responses, with anti-SARS-CoV-2 (N) levels displaying remarkable persistence and anti-SARS-CoV-2 (S) antibodies exhibiting a progressive incline.
Collapse
Affiliation(s)
- Mariam Movsisyan
- Department of Allergology and Clinical Immunology, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
- Cobrain Center, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Nune Truzyan
- Cobrain Center, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Irina Kasparova
- Department of Histology, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Armine Chopikyan
- Cobrain Center, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
- Department of Public Health and Healthcare Organization, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Ra'ed Sawaqed
- General Medicine Faculty, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Alexandra Bedross
- General Medicine Faculty, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Meline Sukiasyan
- Department of Allergology and Clinical Immunology, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
- Cobrain Center, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Karen Dilbaryan
- Cobrain Center, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Sanobar Shariff
- General Medicine Faculty, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Burhan Kantawala
- Cobrain Center, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Gohar Hakobjanyan
- Cobrain Center, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
- Laboratory-Diagnostic Center of Heratsi Clinical Hospital, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Gayane Petrosyan
- Cobrain Center, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
- Laboratory-Diagnostic Center of Heratsi Clinical Hospital, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Armine Hakobyan
- Department of Allergology and Clinical Immunology, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia
| | - Konstantin Yenkoyan
- Neuroscience Laboratory, Cobrain Center, Yerevan State Medical University Named After Mkhitar Heratsi, 0025, Yerevan, Armenia.
- Department of Biochemistry, Yerevan State Medical University Named After Mkhitar Heratsi, Yerevan, Armenia.
| |
Collapse
|
2
|
Limoges MA, Quenum AJI, Chowdhury MMH, Rexhepi F, Namvarpour M, Akbari SA, Rioux-Perreault C, Nandi M, Lucier JF, Lemaire-Paquette S, Premkumar L, Durocher Y, Cantin A, Lévesque S, Dionne IJ, Menendez A, Ilangumaran S, Allard-Chamard H, Piché A, Ramanathan S. SARS-CoV-2 spike antigen-specific B cell and antibody responses in pre-vaccination period COVID-19 convalescent males and females with or without post-covid condition. Front Immunol 2023; 14:1223936. [PMID: 37809081 PMCID: PMC10551145 DOI: 10.3389/fimmu.2023.1223936] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/25/2023] [Indexed: 10/10/2023] Open
Abstract
Background Following SARS-CoV-2 infection a significant proportion of convalescent individuals develop the post-COVID condition (PCC) that is characterized by wide spectrum of symptoms encompassing various organs. Even though the underlying pathophysiology of PCC is not known, detection of viral transcripts and antigens in tissues other than lungs raise the possibility that PCC may be a consequence of aberrant immune response to the viral antigens. To test this hypothesis, we evaluated B cell and antibody responses to the SARS-CoV-2 antigens in PCC patients who experienced mild COVID-19 disease during the pre-vaccination period of COVID-19 pandemic. Methods The study subjects included unvaccinated male and female subjects who developed PCC or not (No-PCC) after clearing RT-PCR confirmed mild COVID-19 infection. SARS-CoV-2 D614G and omicron RBD specific B cell subsets in peripheral circulation were assessed by flow cytometry. IgG, IgG3 and IgA antibody titers toward RBD, spike and nucleocapsid antigens in the plasma were evaluated by ELISA. Results The frequency of the B cells specific to D614G-RBD were comparable in convalescent groups with and without PCC in both males and females. Notably, in females with PCC, the anti-D614G RBD specific double negative (IgD-CD27-) B cells showed significant correlation with the number of symptoms at acute of infection. Anti-spike antibody responses were also higher at 3 months post-infection in females who developed PCC, but not in the male PCC group. On the other hand, the male PCC group also showed consistently high anti-RBD IgG responses compared to all other groups. Conclusions The antibody responses to the spike protein, but not the anti-RBD B cell responses diverge between convalescent males and females who develop PCC. Our findings also suggest that sex-related factors may also be involved in the development of PCC via modulating antibody responses to the SARS-CoV-2 antigens.
Collapse
Affiliation(s)
- Marc-André Limoges
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Sherbrooke, QC, Canada
| | | | | | - Fjolla Rexhepi
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Sherbrooke, QC, Canada
| | - Mozhdeh Namvarpour
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Sherbrooke, QC, Canada
| | - Sara Ali Akbari
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Sherbrooke, QC, Canada
| | - Christine Rioux-Perreault
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Sherbrooke, QC, Canada
| | - Madhuparna Nandi
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Sherbrooke, QC, Canada
| | - Jean-François Lucier
- Department of Biology, Faculty of Science, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Samuel Lemaire-Paquette
- Unité de Recherche Clinique et épidémiologique, Centre de Recherche du CHUS, Sherbrooke, QC, Canada
| | - Lakshmanane Premkumar
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Yves Durocher
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | - André Cantin
- Departments of Medicine, Faculty of Medicine and Health Sciences, Sherbrooke, QC, Canada
| | - Simon Lévesque
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Sherbrooke, QC, Canada
- Laboratoire de Microbiologie, CIUSSS de l’Estrie – CHUS, Sherbrooke, QC, Canada
| | - Isabelle J. Dionne
- Faculty of Physical Activity Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
- Research Centre on Aging, Affiliated with CIUSSS de l’Estrie-CHUS, Sherbrooke, QC, Canada
| | - Alfredo Menendez
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Sherbrooke, QC, Canada
| | - Subburaj Ilangumaran
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Sherbrooke, QC, Canada
| | - Hugues Allard-Chamard
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Alain Piché
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Sherbrooke, QC, Canada
| | - Sheela Ramanathan
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Sherbrooke, QC, Canada
| |
Collapse
|
3
|
Ziganshina MM, Shilova NV, Khalturina EO, Dolgushina NV, V Borisevich S, Yarotskaya EL, Bovin NV, Sukhikh GT. Antibody-Dependent Enhancement with a Focus on SARS-CoV-2 and Anti-Glycan Antibodies. Viruses 2023; 15:1584. [PMID: 37515270 PMCID: PMC10384250 DOI: 10.3390/v15071584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Antibody-dependent enhancement (ADE) is a phenomenon where virus-specific antibodies paradoxically cause enhanced viral replication and/or excessive immune responses, leading to infection exacerbation, tissue damage, and multiple organ failure. ADE has been observed in many viral infections and is supposed to complicate the course of COVID-19. However, the evidence is insufficient. Since no specific laboratory markers have been described, the prediction and confirmation of ADE are very challenging. The only possible predictor is the presence of already existing (after previous infection) antibodies that can bind to viral epitopes and promote the disease enhancement. At the same time, the virus-specific antibodies are also a part of immune response against a pathogen. These opposite effects of antibodies make ADE research controversial. The assignment of immunoglobulins to ADE-associated or virus neutralizing is based on their affinity, avidity, and content in blood. However, these criteria are not clearly defined. Another debatable issue (rather terminological, but no less important) is that in most publications about ADE, all immunoglobulins produced by the immune system against pathogens are qualified as pre-existing antibodies, thus ignoring the conventional use of this term for natural antibodies produced without any stimulation by pathogens. Anti-glycan antibodies (AGA) make up a significant part of the natural immunoglobulins pool, and there is some evidence of their antiviral effect, particularly in COVID-19. AGA have been shown to be involved in ADE in bacterial infections, but their role in the development of ADE in viral infections has not been studied. This review focuses on pros and cons for AGA as an ADE trigger. We also present the results of our pilot studies, suggesting that AGAs, which bind to complex epitopes (glycan plus something else in tight proximity), may be involved in the development of the ADE phenomenon.
Collapse
Affiliation(s)
- Marina M Ziganshina
- National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health of the Russian Federation, Oparina Street 4, 117997 Moscow, Russia
| | - Nadezhda V Shilova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health of the Russian Federation, Oparina Street 4, 117997 Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Eugenia O Khalturina
- National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health of the Russian Federation, Oparina Street 4, 117997 Moscow, Russia
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia
| | - Natalya V Dolgushina
- National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health of the Russian Federation, Oparina Street 4, 117997 Moscow, Russia
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia
| | | | - Ekaterina L Yarotskaya
- National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health of the Russian Federation, Oparina Street 4, 117997 Moscow, Russia
| | - Nicolai V Bovin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Gennady T Sukhikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health of the Russian Federation, Oparina Street 4, 117997 Moscow, Russia
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia
| |
Collapse
|
4
|
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: 0] [Impact Index Per Article: 0] [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
|
5
|
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: 2] [Impact Index Per Article: 2.0] [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
|
6
|
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: 2.0] [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
|
7
|
Fan Q, Nie Z, Xie S. Panorama of Breakthrough Infection Caused by SARS-CoV-2: A Review. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:1733. [PMID: 36556935 PMCID: PMC9784755 DOI: 10.3390/medicina58121733] [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: 10/25/2022] [Revised: 11/22/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022]
Abstract
Since the outbreak of the novel coronavirus disease 2019 (COVID-19) in 2019, many countries have successively developed a variety of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, with the continuous spread of SARS-CoV-2, it has evolved several variants; as a result, prevention and control of the pandemic of SARS-CoV-2 has become more important. Among these variants, the Omicron variant has higher transmissibility and immune escape ability and is the main variant causing a large number of COVID-19 breakthrough infection, thus, presenting new challenges to pandemic prevention and control. Hence, we review the biological characteristics of the Omicron variant and discuss the current status and possible mechanism of breakthrough infection caused by the Omicron variant in order to provide insights into the prevention and control of the pandemic of SARS-CoV-2.
Collapse
Affiliation(s)
| | | | - Songping Xie
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
8
|
Wang K, Luan Z, Guo Z, Ran J, Tian M, Zhao S. The Association Between Clinical Severity and Incubation Period of SARS-CoV-2 Delta Variants: Retrospective Observational Study. JMIR Public Health Surveill 2022; 8:e40751. [PMID: 36346940 PMCID: PMC9678331 DOI: 10.2196/40751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 10/22/2022] [Accepted: 10/27/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND As of August 25, 2021, Jiangsu province experienced the largest COVID-19 outbreak in eastern China that was seeded by SARS-CoV-2 Delta variants. As one of the key epidemiological parameters characterizing the transmission dynamics of COVID-19, the incubation period plays an essential role in informing public health measures for epidemic control. The incubation period of COVID-19 could vary by different age, sex, disease severity, and study settings. However, the impacts of these factors on the incubation period of Delta variants remains uninvestigated. OBJECTIVE The objective of this study is to characterize the incubation period of the Delta variant using detailed contact tracing data. The effects of age, sex, and disease severity on the incubation period were investigated by multivariate regression analysis and subgroup analysis. METHODS We extracted contact tracing data of 353 laboratory-confirmed cases of SARS-CoV-2 Delta variants' infection in Jiangsu province, China, from July to August 2021. The distribution of incubation period of Delta variants was estimated by using likelihood-based approach with adjustment for interval-censored observations. The effects of age, sex, and disease severity on the incubation period were expiated by using multivariate logistic regression model with interval censoring. RESULTS The mean incubation period of the Delta variant was estimated at 6.64 days (95% credible interval: 6.27-7.00). We found that female cases and cases with severe symptoms had relatively longer mean incubation periods than male cases and those with nonsevere symptoms, respectively. One-day increase in the incubation period of Delta variants was associated with a weak decrease in the probability of having severe illness with an adjusted odds ratio of 0.88 (95% credible interval: 0.71-1.07). CONCLUSIONS In this study, the incubation period was found to vary across different levels of sex, age, and disease severity of COVID-19. These findings provide additional information on the incubation period of Delta variants and highlight the importance of continuing surveillance and monitoring of the epidemiological characteristics of emerging SARS-CoV-2 variants as they evolve.
Collapse
Affiliation(s)
- Kai Wang
- Department of Medical Engineering and Technology, Xinjiang Medical University, Urumqi, China
| | - Zemin Luan
- Department of Medical Engineering and Technology, Xinjiang Medical University, Urumqi, China
| | - Zihao Guo
- JC School of Public Health and Primary Care, Chinese University of Hong Kong, Hong Kong, China (Hong Kong)
| | - Jinjun Ran
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Maozai Tian
- Department of Medical Engineering and Technology, Xinjiang Medical University, Urumqi, China
| | - Shi Zhao
- JC School of Public Health and Primary Care, Chinese University of Hong Kong, Hong Kong, China (Hong Kong)
| |
Collapse
|
9
|
Chen S, Guan F, Candotti F, Benlagha K, Camara NOS, Herrada AA, James LK, Lei J, Miller H, Kubo M, Ning Q, Liu C. The role of B cells in COVID-19 infection and vaccination. Front Immunol 2022; 13:988536. [PMID: 36110861 PMCID: PMC9468879 DOI: 10.3389/fimmu.2022.988536] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 07/26/2022] [Indexed: 12/23/2022] Open
Abstract
B cells secrete antibodies and mediate the humoral immune response, making them extremely important in protective immunity against SARS-CoV-2, which caused the coronavirus disease 2019 (COVID-19) pandemic. In this review, we summarize the positive function and pathological response of B cells in SARS-CoV-2 infection and re-infection. Then, we structure the immunity responses that B cells mediated in peripheral tissues. Furthermore, we discuss the role of B cells during vaccination including the effectiveness of antibodies and memory B cells, viral evolution mechanisms, and future vaccine development. This review might help medical workers and researchers to have a better understanding of the interaction between B cells and SARS-CoV-2 and broaden their vision for future investigations.
Collapse
Affiliation(s)
- Shiru Chen
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science Technology, Wuhan, China
- Department of Internal Medicine, The Division of Gastroenterology and Hepatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Guan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science Technology, Wuhan, China
| | - Fabio Candotti
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Kamel Benlagha
- Institut de Recherche Saint-Louis, Université de Paris, Paris, France
| | - Niels Olsen Saraiva Camara
- Laboratory of Human Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Andres A. Herrada
- Lymphatic and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomedicas, Universidad Autonoma de Chile, Talca, Chile
| | - Louisa K. James
- Centre for Immunobiology, Bizard Institute, Queen Mary University of London, London, United Kingdom
| | - Jiahui Lei
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science Technology, Wuhan, China
| | - Heather Miller
- Cytek Biosciences, R&D Clinical Reagents, Fremont, CA, United States
| | - Masato Kubo
- Laboratory for Cytokine Regulation, Center for Integrative Medical Science (IMS), Rikagaku Kenkyusho, Institute of Physical and Chemical Research (RIKEN) Yokohama Institute, Yokohama, Kanagawa, Japan
| | - Qin Ning
- Department of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science Technology, Wuhan, China
| |
Collapse
|
10
|
Colton H, Hodgson D, Hornsby H, Brown R, Mckenzie J, Bradley KL, James C, Lindsey BB, Birch S, Marsh L, Wood S, Bayley M, Dickson G, James DC, Nicklin MJ, Sayers JR, Zafred D, Rowland-Jones SL, Kudesia G, Kucharski A, Darton TC, de Silva TI, Collini PJ. Risk factors for SARS-CoV-2 seroprevalence following the first pandemic wave in UK healthcare workers in a large NHS Foundation Trust. Wellcome Open Res 2022; 6:220. [PMID: 35600250 PMCID: PMC9091808 DOI: 10.12688/wellcomeopenres.17143.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2022] [Indexed: 11/22/2022] Open
Abstract
Background: We aimed to measure SARS-CoV-2 seroprevalence in a cohort of healthcare workers (HCWs) during the first UK wave of the COVID-19 pandemic, explore risk factors associated with infection, and investigate the impact of antibody titres on assay sensitivity. Methods: HCWs at Sheffield Teaching Hospitals NHS Foundation Trust were prospectively enrolled and sampled at two time points. We developed an in-house ELISA for testing participant serum for SARS-CoV-2 IgG and IgA reactivity against Spike and Nucleoprotein. Data were analysed using three statistical models: a seroprevalence model, an antibody kinetics model, and a heterogeneous sensitivity model. Results: Our in-house assay had a sensitivity of 99·47% and specificity of 99·56%. We found that 24·4% (n=311/1275) of HCWs were seropositive as of 12th June 2020. Of these, 39·2% (n=122/311) were asymptomatic. The highest adjusted seroprevalence was measured in HCWs on the Acute Medical Unit (41·1%, 95% CrI 30·0-52·9) and in Physiotherapists and Occupational Therapists (39·2%, 95% CrI 24·4-56·5). Older age groups showed overall higher median antibody titres. Further modelling suggests that, for a serological assay with an overall sensitivity of 80%, antibody titres may be markedly affected by differences in age, with sensitivity estimates of 89% in those over 60 years but 61% in those ≤30 years. Conclusions: HCWs in acute medical units and those working closely with COVID-19 patients were at highest risk of infection, though whether these are infections acquired from patients or other staff is unknown. Current serological assays may underestimate seroprevalence in younger age groups if validated using sera from older and/or more severe COVID-19 cases.
Collapse
Affiliation(s)
- Hayley Colton
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - David Hodgson
- Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Hailey Hornsby
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Rebecca Brown
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Joanne Mckenzie
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Kirsty L. Bradley
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Cameron James
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Benjamin B. Lindsey
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Sarah Birch
- Academic Directorate of Communicable Diseases and Specialised Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
| | - Louise Marsh
- Academic Directorate of Communicable Diseases and Specialised Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
| | - Steven Wood
- Department of Scientific Computing and Informatics, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
| | - Martin Bayley
- Department of Scientific Computing and Informatics, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
| | - Gary Dickson
- Department of Scientific Computing and Informatics, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
| | - David C. James
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, S10 2TN, UK
| | - Martin J. Nicklin
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Jon R. Sayers
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, S10 2TN, UK
- Sheffield Institute for Nucleic Acids, University of Sheffield, Sheffield, S10 2TN, UK
| | - Domen Zafred
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Sarah L. Rowland-Jones
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, S10 2TN, UK
| | - Goura Kudesia
- Department of Virology, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S5 7AU, UK
| | - Adam Kucharski
- Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - CMMID COVID-19 Working Group
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
- Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
- Academic Directorate of Communicable Diseases and Specialised Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Scientific Computing and Informatics, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, S10 2TN, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, S10 2TN, UK
- Sheffield Institute for Nucleic Acids, University of Sheffield, Sheffield, S10 2TN, UK
- Department of Virology, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S5 7AU, UK
| | - Thomas C. Darton
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, S10 2TN, UK
| | - Thushan I. de Silva
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, S10 2TN, UK
| | - Paul J. Collini
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, S10 2TN, UK
| |
Collapse
|
11
|
Low SARS-CoV-2 antibody titers may be associated with poor clinical outcomes for patients with severe COVID-19. Sci Rep 2022; 12:9147. [PMID: 35650227 PMCID: PMC9159042 DOI: 10.1038/s41598-022-12834-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 05/17/2022] [Indexed: 12/14/2022] Open
Abstract
Recently, immune response to coronavirus disease (COVID-19) has attracted attention where an association between higher antibody titer and worsening disease severity has been reported. However, our experiences with severe COVID-19 patients with low antibody titers led to hypothesizing that suppressed humoral immune response may be associated with poorer prognosis in severe COVID19. In this study, antibody titers in severe COVID19 patients were measured at 7, 10, 12, and 14 days after onset. Patients were divided into survivors and non-survivors. SARS-CoV-2 IgM in survivors and non-survivors were 0.06 AU and 0.02 AU (P = 0.048) at 10 days, 0.1 AU and 0.03 AU (P = 0.02) at 12 days, and 0.17 AU and 0.06 AU (P = 0.02) at 14 days. IgG in survivors and non-survivors were 0.01 AU and 0.01 AU (P = 0.04) at 7 days, 0.42 AU and 0.01 AU (P = 0.04) at 12 days, and 0.42 AU and 0.01 AU (P = 0.02) at 14 days. Multivariate analysis showed better survival among patients with IgM positivity at 12 days (P = 0.04), IgG positivity at 12 days (P = 0.04), IgM positivity at 14 days (P = 0.008), and IgG positivity at 14 days (P = 0.005). In severe COVID-19, low antibody titers on days 12 and 14 after onset were associated with poorer prognosis.
Collapse
|
12
|
Puzyrenko A, Felix JC, Ledeboer NA, Sun Y, Rui H, Sheinin Y. Cytotoxic CD8-positive T-lymphocyte infiltration in the lungs as a histological pattern of SARS-CoV-2 pneumonitis. Pathology 2022; 54:404-408. [PMID: 34836647 PMCID: PMC8572726 DOI: 10.1016/j.pathol.2021.09.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/05/2021] [Accepted: 09/06/2021] [Indexed: 11/05/2022]
Abstract
Despite millions of PCR confirmed cases of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection, the long-term pathophysiological changes induced by this infection in the lungs and their relationship with possible immune triggers remain incompletely understood. Acute respiratory distress syndrome and subsequent respiratory failure are the most common causes of mortality in hospitalised patients. Severe lung tissue destruction can be due to an overactive immune system that far exceeds the harm that would have been caused by direct virus replication. This study extends our previous investigation and presents detailed histopathological findings on cryotransbronchial biopsy in patients with persistent (range 31-182 days) pneumonitis and severe interstitial inflammatory infiltration in the lungs due to SARS-CoV-2 infection. We describe a novel lung injury pattern associated with SARS-CoV-2 pneumonitis, which manifests as a marked interstitial CD8-positive T-cell lymphocytic infiltration. These findings provide a better understanding of the changes in the lungs that ensue due to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Andrii Puzyrenko
- Medical College of Wisconsin, Pathology Department, Milwaukee, WI, USA.
| | - Juan C Felix
- Medical College of Wisconsin, Pathology Department, Milwaukee, WI, USA
| | - Nathan A Ledeboer
- Medical College of Wisconsin, Pathology Department, Milwaukee, WI, USA
| | - Yunguang Sun
- Medical College of Wisconsin, Pathology Department, Milwaukee, WI, USA
| | - Hallgeir Rui
- Medical College of Wisconsin, Pathology Department, Milwaukee, WI, USA
| | - Yuri Sheinin
- Medical College of Wisconsin, Pathology Department, Milwaukee, WI, USA
| |
Collapse
|
13
|
Farouq MAH, Acevedo R, Ferro VA, Mulheran PA, Al Qaraghuli MM. The Role of Antibodies in the Treatment of SARS-CoV-2 Virus Infection, and Evaluating Their Contribution to Antibody-Dependent Enhancement of Infection. Int J Mol Sci 2022; 23:6078. [PMID: 35682757 PMCID: PMC9181534 DOI: 10.3390/ijms23116078] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/26/2022] [Accepted: 05/26/2022] [Indexed: 02/06/2023] Open
Abstract
Antibodies play a crucial role in the immune response, in fighting off pathogens as well as helping create strong immunological memory. Antibody-dependent enhancement (ADE) occurs when non-neutralising antibodies recognise and bind to a pathogen, but are unable to prevent infection, and is widely known and is reported as occurring in infection caused by several viruses. This narrative review explores the ADE phenomenon, its occurrence in viral infections and evaluates its role in infection by SARS-CoV-2 virus, which causes coronavirus disease 2019 (COVID-19). As of yet, there is no clear evidence of ADE in SARS-CoV-2, though this area is still subject to further study.
Collapse
Affiliation(s)
- Mohammed A. H. Farouq
- Department of Chemical and Process Engineering, University of Strathclyde, 75 Montrose Street, Glasgow G1 1XJ, UK; (P.A.M.); (M.M.A.Q.)
| | - Reinaldo Acevedo
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK;
| | - Valerie A. Ferro
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK;
| | - Paul A. Mulheran
- Department of Chemical and Process Engineering, University of Strathclyde, 75 Montrose Street, Glasgow G1 1XJ, UK; (P.A.M.); (M.M.A.Q.)
| | - Mohammed M. Al Qaraghuli
- Department of Chemical and Process Engineering, University of Strathclyde, 75 Montrose Street, Glasgow G1 1XJ, UK; (P.A.M.); (M.M.A.Q.)
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK;
- EPSRC Future Manufacturing Research Hub for Continuous Manufacturing and Advanced Crystallisation (CMAC), University of Strathclyde, 99 George Street, Glasgow G1 1RD, UK
| |
Collapse
|
14
|
Colton H, Hodgson D, Hornsby H, Brown R, Mckenzie J, Bradley KL, James C, Lindsey BB, Birch S, Marsh L, Wood S, Bayley M, Dickson G, James DC, Nicklin MJ, Sayers JR, Zafred D, Rowland-Jones SL, Kudesia G, Kucharski A, Darton TC, de Silva TI, Collini PJ. Risk factors for SARS-CoV-2 seroprevalence following the first pandemic wave in UK healthcare workers in a large NHS Foundation Trust. Wellcome Open Res 2022; 6:220. [PMID: 35600250 PMCID: PMC9091808 DOI: 10.12688/wellcomeopenres.17143.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2022] [Indexed: 11/20/2022] Open
Abstract
Background: We aimed to measure SARS-CoV-2 seroprevalence in a cohort of healthcare workers (HCWs) during the first UK wave of the COVID-19 pandemic, explore risk factors associated with infection, and investigate the impact of antibody titres on assay sensitivity. Methods: HCWs at Sheffield Teaching Hospitals NHS Foundation Trust were prospectively enrolled and sampled at two time points. We developed an in-house ELISA for testing participant serum for SARS-CoV-2 IgG and IgA reactivity against Spike and Nucleoprotein. Data were analysed using three statistical models: a seroprevalence model, an antibody kinetics model, and a heterogeneous sensitivity model. Results: Our in-house assay had a sensitivity of 99·47% and specificity of 99·56%. We found that 24·4% (n=311/1275) of HCWs were seropositive as of 12th June 2020. Of these, 39·2% (n=122/311) were asymptomatic. The highest adjusted seroprevalence was measured in HCWs on the Acute Medical Unit (41·1%, 95% CrI 30·0-52·9) and in Physiotherapists and Occupational Therapists (39·2%, 95% CrI 24·4-56·5). Older age groups showed overall higher median antibody titres. Further modelling suggests that, for a serological assay with an overall sensitivity of 80%, antibody titres may be markedly affected by differences in age, with sensitivity estimates of 89% in those over 60 years but 61% in those ≤30 years. Conclusions: HCWs in acute medical units and those working closely with COVID-19 patients were at highest risk of infection, though whether these are infections acquired from patients or other staff is unknown. Current serological assays may underestimate seroprevalence in younger age groups if validated using sera from older and/or more severe COVID-19 cases.
Collapse
Affiliation(s)
- Hayley Colton
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - David Hodgson
- Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Hailey Hornsby
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Rebecca Brown
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Joanne Mckenzie
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Kirsty L. Bradley
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Cameron James
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Benjamin B. Lindsey
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Sarah Birch
- Academic Directorate of Communicable Diseases and Specialised Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
| | - Louise Marsh
- Academic Directorate of Communicable Diseases and Specialised Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
| | - Steven Wood
- Department of Scientific Computing and Informatics, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
| | - Martin Bayley
- Department of Scientific Computing and Informatics, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
| | - Gary Dickson
- Department of Scientific Computing and Informatics, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
| | - David C. James
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, S10 2TN, UK
| | - Martin J. Nicklin
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Jon R. Sayers
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, S10 2TN, UK
- Sheffield Institute for Nucleic Acids, University of Sheffield, Sheffield, S10 2TN, UK
| | - Domen Zafred
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Sarah L. Rowland-Jones
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, S10 2TN, UK
| | - Goura Kudesia
- Department of Virology, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S5 7AU, UK
| | - Adam Kucharski
- Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - CMMID COVID-19 Working Group
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
- Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
- Academic Directorate of Communicable Diseases and Specialised Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Scientific Computing and Informatics, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, S10 2TN, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, S10 2TN, UK
- Sheffield Institute for Nucleic Acids, University of Sheffield, Sheffield, S10 2TN, UK
- Department of Virology, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S5 7AU, UK
| | - Thomas C. Darton
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, S10 2TN, UK
| | - Thushan I. de Silva
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, S10 2TN, UK
| | - Paul J. Collini
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, S10 2TN, UK
| |
Collapse
|
15
|
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: 17.0] [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
|
16
|
Abstract
The process of adaptation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to humans probably had started decades ago, when its ancestor diverged from the bat coronavirus. The adaptive process comprises strategies the virus uses to overcome the respiratory tract defense barriers and replicate and shed in the host cells. These strategies include the impairment of interferon production, hiding immunogenic motifs, avoiding viral RNA detection, manipulating cell autophagy, triggering host cell death, inducing lymphocyte exhaustion and depletion, and finally, mutation and escape from immunity. In addition, SARS-CoV-2 employs strategies to take advantage of host cell resources for its benefits, such as inhibiting the ubiquitin-proteasome system, hijacking mitochondria functions, and usage of enhancing antibodies. It may be anticipated that as the tradeoffs of adaptation progress, the virus destructive burden will gradually subside. Some evidence suggests that SARS-CoV-2 will become part of the human respiratory virome, as had occurred with other coronaviruses, and coevolve with its host.
Collapse
Affiliation(s)
- Eduardo Tosta
- Universidade de Brasília, Faculdade de Medicina, Brasília, DF, Brasil
| |
Collapse
|
17
|
Moorthy M, Miraclin A, Thomas J, John J, Kandoth Y, Livingstone J, Wilson B, Vanjare H, Jose A, Zachariah A, Sivadasan A. COVID-19 associated parainfectious encephalomyelitis: A unique case with early cns immunological response and practical implications. Ann Indian Acad Neurol 2022; 25:167-169. [PMID: 35342265 PMCID: PMC8954339 DOI: 10.4103/aian.aian_29_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/05/2021] [Accepted: 04/21/2021] [Indexed: 11/23/2022] Open
|
18
|
Wu D, Zhang X, Ziemba Y, Haghi N, Brody J, Hsu P. Dynamics of Peripheral Blood T-lymphocytes Have Predictive Values for the Clinical Outcome of COVID-19 Patients in Intensive Care Unit. CLINICAL PATHOLOGY (THOUSAND OAKS, VENTURA COUNTY, CALIF.) 2022; 15:2632010X211072818. [PMID: 35083433 PMCID: PMC8785304 DOI: 10.1177/2632010x211072818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 12/14/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) patients with severe disease had a high mortality rate. It's imperative to identify risk factors associated with disease progression and prognosis. Immune responses played an important role in the host's defense against the virus. We studied the dynamics of peripheral blood lymphocytes (PBLs) in relation to the clinical outcome in COVID-19 patients in intensive care unit (ICU). DESIGN This cohort included 342 COVID-19 patients who were admitted to ICU between February 1 and May 30, 2020, with 178 having follow-up PBL analysis. The patients were divided into a group that survived and an expired group. PBL analysis was performed by flow cytometry. RESULTS At time of initial flow analysis, there were no statistically significant differences in lymphocyte, T-cell and subsets, B-cell or natural killer (NK) cell counts between the 2 groups. However, during the ICU course, the surviving group demonstrated a full recovery of CD3+ T-cells, CD4+ T-cells, and CD8+ T-cells, with no significant change in B-cells, and a slight upward trend in NK-cells. In contrast, the expired group showed no recovery in T-cells (and subsets) and no significant changes in B-cells and NK-cells. We identified the earliest time points and cut-off values for T-cell subsets that predict clinical outcomes. CONCLUSION The results of this study suggest that evaluation of PBL in COVID-19 patients could be valuable in the study of the immune responses to the disease and the prognostication of outcome.
Collapse
Affiliation(s)
- Dongling Wu
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Xinmin Zhang
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Yonah Ziemba
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Nina Haghi
- Department of Pathology and Laboratory Medicine, NYU Langone Health, New York, NY, USA
| | - Judith Brody
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Peihong Hsu
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| |
Collapse
|
19
|
Kundu A, Maji S, Kumar S, Bhattacharya S, Chakraborty P, Sarkar J. Clinical aspects and presumed etiology of multisystem inflammatory syndrome in children (MIS-C): A review. CLINICAL EPIDEMIOLOGY AND GLOBAL HEALTH 2022; 14:100966. [PMID: 35132389 PMCID: PMC8810427 DOI: 10.1016/j.cegh.2022.100966] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/08/2022] [Accepted: 01/12/2022] [Indexed: 12/21/2022] Open
Abstract
The COVID-19 outbreak sparked by SARS-CoV-2, begat significant rates of malady worldwide, where children with an abnormal post-COVID ailment called the Multisystem Inflammatory Syndrome (MIS-C), were reported by April 2020. Here we have reviewed the clinical characteristics of the pediatric patients and the prognosis currently being utilized. A vivid comparison of MIS-C with other clinical conditions has been done. We have addressed the probable etiology and fundamental machinery of the inflammatory reactions, which drive organ failure. The involvement of androgen receptors portrays the likelihood of asymptomatic illness in children below adolescence, contributing to the concept of antibody-dependent enhancement.
Collapse
Key Words
- ACE2, Angiotensin-Converting Enzyme-2
- ADE, Antibody-Dependent Enhancement
- AR, Allosomal Androgen Receptor
- ARDS, Acute Respiratory Distress Syndrome
- BNP, Brain Natriuretic Peptide
- CDC, Centres for Disease Control and Prevention
- CRP, C-reactive protein
- ESR, Erythrocyte Sedimentation Rate
- IVIG, Intravenous Immunoglobulin
- KD, Kawasaki Disease
- Kawasaki disease
- LVEF, Left Ventricular Ejection Fraction
- MIS-C
- MIS-C, Multisystem Inflammatory Syndrome in Children
- Macrophage and antibody-dependent enhancement (ADE)
- Multiorgan failure
- NLRP3, NLR family Pyrin Domain Containing 3
- PCAID, Pediatric COVID-19 Associated Inflammatory Disorder
- PIMS-TS, Pediatric Inflammatory Multisystem Syndrome Temporally Associated
- PPT, Prolonged Prothrombin Time
- PTT, The Prothrombin Time Test
- Pediatric patient
- RT-PCR, Real Time- Polymerase Chain Reaction
- SARS-COV-2, Severe Acute Respiratory Syndrome Coronavirus 2
- SARS-CoV-2
- SHLH/MAS, Secondary Hemophagocytic Lymphohistiocytosis/Macrophage Activation Syndrome
- TMPRSS2, Transmembrane Protease, Serine 2
- TNP, Tumour Necrosis Factor
- TSS, Toxic Shock Syndrome
- TTSPs, Type II Transmembrane Serine Protease
Collapse
Affiliation(s)
- Anusrita Kundu
- Department of Botany, Bethune College, Manicktala, Kolkata, West Bengal, 700006, India
| | - Swagata Maji
- Department of Botany, Bethune College, Manicktala, Kolkata, West Bengal, 700006, India
| | - Suchismita Kumar
- Department of Botany, Bethune College, Manicktala, Kolkata, West Bengal, 700006, India
| | - Shreya Bhattacharya
- Department of Botany, Bethune College, Manicktala, Kolkata, West Bengal, 700006, India
| | - Pallab Chakraborty
- Department of Botany, Acharya Prafulla Chandra College, New Barrakpur, Kolkata, West Bengal, 700131, India
| | - Joy Sarkar
- Department of Botany, Dinabandhu Andrews College, Garia, Kolkata, West Bengal, 700084, India,Corresponding author
| |
Collapse
|
20
|
Alefishat E, Jelinek HF, Mousa M, Tay GK, Alsafar HS. Immune response to SARS-CoV-2 Variants: A focus on severity, susceptibility, and preexisting immunity. J Infect Public Health 2022; 15:277-288. [PMID: 35074728 PMCID: PMC8757655 DOI: 10.1016/j.jiph.2022.01.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/16/2021] [Accepted: 01/09/2022] [Indexed: 01/08/2023] Open
Abstract
The heterogeneous phenotypes among patients with coronavirus disease 2019 (COVID-19) has drawn worldwide attention, especially those with severe symptoms without comorbid conditions. Immune responses to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the causative virus of COVID-19, occur mainly by the innate immune response via the interferon (IFN)-mediated pathways, and the adaptive immunity via the T lymphocyte and the antibody mediated pathways. The ability of the original Wuhan SARS-CoV-2 strain, and possibly more so with new emerging variants, to antagonize IFN-mediated antiviral responses can be behind the higher early viral load, higher transmissibility, and milder symptoms compared to SARS-CoV and are part of the continued clinical evolution of COVID-19. Since it first emerged, several variants of SARS-CoV-2 have been circulating worldwide. Variants that have the potential to elude natural or vaccine-mediated immunity are variants of concern. This review focuses on the main host factors that may explain the immune responses to SARS-CoV-2 and its variants in the context of susceptibility, severity, and preexisting immunity.
Collapse
Affiliation(s)
- Eman Alefishat
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Department of Pharmacology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Department of Biopharmaceutics and Clinical Pharmacy, Faculty of Pharmacy, The University of Jordan, Amman, Jordan
| | - Herbert F Jelinek
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Department of Biomedical Engineering, College of Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Center of Heath Engineering Innovation, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Mira Mousa
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Nuffield Department of Women's and Reproduction Health, Oxford University, Oxford, United Kingdom
| | - Guan K Tay
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Discipline of Psychiatry, Medical School, the University of Western Australia, Perth WA, Australia; School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Habiba S Alsafar
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Department of Biomedical Engineering, College of Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Department of Genetics and Molecular Biology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Emirates Bio-Research Center, Ministry of Interior, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
21
|
Epidemiological features of COVID-19 patients with prolonged incubation period and its implications for controlling the epidemics in China. BMC Public Health 2021; 21:2239. [PMID: 34886835 PMCID: PMC8655494 DOI: 10.1186/s12889-021-12337-9] [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: 07/28/2021] [Accepted: 11/29/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND COVID-19 patients with long incubation period were reported in clinical practice and tracing of close contacts, but their epidemiological or clinical features remained vague. METHODS We analyzed 11,425 COVID-19 cases reported between January-August, 2020 in China. The accelerated failure time model, Logistic and modified Poisson regression models were used to investigate the determinants of prolonged incubation period, as well as their association with clinical severity and transmissibility, respectively. RESULT Among local cases, 268 (10.2%) had a prolonged incubation period of > 14 days, which was more frequently seen among elderly patients, those residing in South China, with disease onset after Level I response measures administration, or being exposed in public places. Patients with prolonged incubation period had lower risk of severe illness (ORadjusted = 0.386, 95% CI: 0.203-0.677). A reduced transmissibility was observed for the primary patients with prolonged incubation period (50.4, 95% CI: 32.3-78.6%) than those with an incubation period of ≤14 days. CONCLUSIONS The study provides evidence supporting a prolonged incubation period that exceeded 2 weeks in over 10% for COVID-19. Longer monitoring periods than 14 days for quarantine or persons potentially exposed to SARS-CoV-2 should be justified in extreme cases, especially for those elderly.
Collapse
|
22
|
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
|
23
|
SARS-CoV-2 Neutralizing Antibody Levels Post COVID-19 Vaccination Based on ELISA Method-A Small Real-World Sample Exploration. Vaccines (Basel) 2021; 9:vaccines9101139. [PMID: 34696247 PMCID: PMC8541171 DOI: 10.3390/vaccines9101139] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/17/2021] [Accepted: 09/28/2021] [Indexed: 11/17/2022] Open
Abstract
This study investigated the detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) neutralizing antibodies following inoculation with the coronavirus disease (COVID-19) vaccine. From June to July 2021, 127 participants who had completed COVID-19 vaccination (inactivated SARS-CoV-2 vaccine, 64; CoronaVac, 61; CanSino, 2) were recruited and tested using SARS-CoV-2 neutralizing antibody kits. The positive detection rate (inhibition of neutralizing antibodies ≥ 30%) was calculated and stratified according to population characteristics and inoculation time. The positive rate of neutralizing antibody was 47.22% (17/36) in men and 53.85% (49/91) in women, and 54.55% (24/44) in BMI ≥ 24 and 50.60% (42/83) in BMI < 24. Age was stratified as 20-29, 30-39, 40-49, and ≥50; positive detection rates of SARS-CoV-2 neutralizing antibodies were observed in 60.00% (24/40), 50.00% (21/42), 48.39% (15/31), and 42.86% (6/14), respectively, but with no significant difference (x2 = 1.724, p = 0.632). Among 127 vaccinated participants, 66 (51.97%) were positive. The positive detection rate was 63.93% (39/61) with CoronaVac and 42.19% (27/64) with the inactivated SARS-CoV-2 vaccine (significance x2 = 5.927, p = 0.015). Multivariate analysis revealed a significant difference in vaccination times, with average vaccination weeks in the positive and negative groups of 11.57 ± 6.48 and 17.87 ± 9.17, respectively (t= -4.501, p < 0.001). The positive neutralizing antibody rate was 100.00%, 60.00%, 58.33%, 55.56%, 43.14%, 28.57%, and 0.00% at 2-4, 5-8, 9-12, 13-16,17-20, 21-24, and >24 weeks, respectively (x2 = 18.030, p = 0.006). Neutralizing antibodies were detected after COVID-19 inoculation, with differences relating to inoculation timing. This study provides a reference for vaccine evaluation and follow-up immunization strengthening.
Collapse
|
24
|
Colton H, Hodgson D, Hornsby H, Brown R, Mckenzie J, Bradley KL, James C, Lindsey BB, Birch S, Marsh L, Wood S, Bayley M, Dickson G, James DC, Nicklin MJ, Sayers JR, Zafred D, Rowland-Jones SL, Kudesia G, Kucharski A, Darton TC, de Silva TI, Collini PJ. Risk factors for SARS-CoV-2 seroprevalence following the first pandemic wave in UK healthcare workers in a large NHS Foundation Trust. Wellcome Open Res 2021; 6:220. [PMID: 35600250 PMCID: PMC9091808 DOI: 10.12688/wellcomeopenres.17143.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2021] [Indexed: 12/23/2022] Open
Abstract
Background: We aimed to measure SARS-CoV-2 seroprevalence in a cohort of healthcare workers (HCWs) during the first UK wave of the COVID-19 pandemic, explore risk factors associated with infection, and investigate the impact of antibody titres on assay sensitivity. Methods: HCWs at Sheffield Teaching Hospitals NHS Foundation Trust were prospectively enrolled and sampled at two time points. SARS-CoV-2 antibodies were tested using an in-house assay for IgG and IgA reactivity against Spike and Nucleoprotein (sensitivity 99·47%, specificity 99·56%). Data were analysed using three statistical models: a seroprevalence model, an antibody kinetics model, and a heterogeneous sensitivity model. Results: As of 12th June 2020, 24·4% (n=311/1275) of HCWs were seropositive. Of these, 39·2% (n=122/311) were asymptomatic. The highest adjusted seroprevalence was measured in HCWs on the Acute Medical Unit (41·1%, 95% CrI 30·0-52·9) and in Physiotherapists and Occupational Therapists (39·2%, 95% CrI 24·4-56·5). Older age groups showed overall higher median antibody titres. Further modelling suggests that, for a serological assay with an overall sensitivity of 80%, antibody titres may be markedly affected by differences in age, with sensitivity estimates of 89% in those over 60 years but 61% in those ≤30 years. Conclusions: HCWs in acute medical units working closely with COVID-19 patients were at highest risk of infection, though whether these are infections acquired from patients or other staff is unknown. Current serological assays may underestimate seroprevalence in younger age groups if validated using sera from older and/or more symptomatic individuals.
Collapse
Affiliation(s)
- Hayley Colton
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - David Hodgson
- Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Hailey Hornsby
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Rebecca Brown
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Joanne Mckenzie
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Kirsty L. Bradley
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Cameron James
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Benjamin B. Lindsey
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Sarah Birch
- Academic Directorate of Communicable Diseases and Specialised Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
| | - Louise Marsh
- Academic Directorate of Communicable Diseases and Specialised Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
| | - Steven Wood
- Department of Scientific Computing and Informatics, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
| | - Martin Bayley
- Department of Scientific Computing and Informatics, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
| | - Gary Dickson
- Department of Scientific Computing and Informatics, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
| | - David C. James
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, S10 2TN, UK
| | - Martin J. Nicklin
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Jon R. Sayers
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, S10 2TN, UK
- Sheffield Institute for Nucleic Acids, University of Sheffield, Sheffield, S10 2TN, UK
| | - Domen Zafred
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Sarah L. Rowland-Jones
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, S10 2TN, UK
| | - Goura Kudesia
- Department of Virology, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S5 7AU, UK
| | - Adam Kucharski
- Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - CMMID COVID-19 Working Group
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
- Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
- Academic Directorate of Communicable Diseases and Specialised Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Scientific Computing and Informatics, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, S10 2TN, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, S10 2TN, UK
- Sheffield Institute for Nucleic Acids, University of Sheffield, Sheffield, S10 2TN, UK
- Department of Virology, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S5 7AU, UK
| | - Thomas C. Darton
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, S10 2TN, UK
| | - Thushan I. de Silva
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, S10 2TN, UK
| | - Paul J. Collini
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, S10 2JF, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, S10 2TN, UK
| |
Collapse
|
25
|
Hosseini SA, Zahedipour F, Mirzaei H, Kazemi Oskuee R. Potential SARS-CoV-2 vaccines: Concept, progress, and challenges. Int Immunopharmacol 2021; 97:107622. [PMID: 33895475 PMCID: PMC8006194 DOI: 10.1016/j.intimp.2021.107622] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/25/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023]
Abstract
Since September 2020, the world has had more than 28 million cases of coronavirus disease 2019 (COVID-19). Many countries are facing a second wave of the COVID-19 outbreak. A pressing need is evident for the development of a potent vaccine to control the SARS-CoV-2. Institutions and companies in many countries have announced their vaccine research programs and progress against the COVID-19. While most vaccines go through the designation and preparation stages, some of them are under evaluation for efficacy among animal models and clinical trials, and three approved vaccine candidates have been introduced for limited exploitation in Russia and China. An effective vaccine must induce a protective response of both cell-mediated and humoral immunity and should meet the safety and efficacy criteria. Although the emergence of new technologies has accelerated the development of vaccines, there are several challenges on the way, such as limited knowledge about the pathophysiology of the virus, inducing humoral or cellular immunity, immune enhancement with animal coronavirus vaccines, and lack of an appropriate animal model. In this review, we firstly discuss the immune responses against SARS-CoV-2 disease, subsequently, give an overview of several vaccine platforms for SARS-CoV-2 under clinical trials and challenges in vaccine development against this virus.
Collapse
Affiliation(s)
- Seyede Atefe Hosseini
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran,Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Zahedipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran,Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran,Corresponding authors
| | - Reza Kazemi Oskuee
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran,Corresponding authors
| |
Collapse
|
26
|
Chen J, Liu X, Zhang X, Lin Y, Liu D, Xun J, Wang Z, Gu L, Li Q, Yin D, Yang J, Lu H. Decline in neutralising antibody responses, but sustained T-cell immunity, in COVID-19 patients at 7 months post-infection. Clin Transl Immunology 2021; 10:e1319. [PMID: 34336207 PMCID: PMC8313961 DOI: 10.1002/cti2.1319] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/27/2021] [Accepted: 07/07/2021] [Indexed: 12/26/2022] Open
Abstract
Objectives This study aimed to explore the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific humoral responses and T-cell responses in patients who have recovered from coronavirus disease 2019 (COVID-19) to understand the natural protective immune responses and to facilitate the development of vaccines. Methods We conducted a combined assessment of the changes in neutralising antibody levels and SARS-CoV-2-specific T-cell responses over time in 27 patients up to 7 months after infection. Results The neutralising antibody remained detectable in 96.3% of the patients at their second visit at about 7 months post-onset of symptoms. However, their humoral responses, including titres of the spike receptor-binding domain IgG and neutralising antibody, decreased significantly compared with those at first clinic visit. By contrast, the proportions of spike-specific CD4+ T cells, but not CD8+ T cells, in COVID-19 patients after recovery were persistently higher than those in healthy controls. No significant change was observed in the proportion of spike-specific CD4+ T cells in patients who had recovered from COVID-19 within 7 months. Conclusion The SARS-CoV-2-specific T-cell immune responses persisted, while the neutralising antibodies decayed. Further studies are needed to extend the longevity of neutralising antibodies and to evaluate whether these T cells are sufficient to protect patients from reinfection.
Collapse
Affiliation(s)
- Jun Chen
- Department of Infectious Diseases and Immunology Shanghai Public Health Clinical Center Fudan University Shanghai China
| | - Xiaomin Liu
- Scientific Research Center Shanghai Public Health Clinical Center Fudan University Shanghai China
| | - Xinyu Zhang
- Scientific Research Center Shanghai Public Health Clinical Center Fudan University Shanghai China
| | - Yixiao Lin
- Department of Infectious Diseases and Immunology Shanghai Public Health Clinical Center Fudan University Shanghai China
| | - Danping Liu
- Scientific Research Center Shanghai Public Health Clinical Center Fudan University Shanghai China
| | - Jingna Xun
- Scientific Research Center Shanghai Public Health Clinical Center Fudan University Shanghai China
| | - Zhenyan Wang
- Department of Infectious Diseases and Immunology Shanghai Public Health Clinical Center Fudan University Shanghai China
| | - Ling Gu
- Scientific Research Center Shanghai Public Health Clinical Center Fudan University Shanghai China
| | - Qian Li
- Scientific Research Center Shanghai Public Health Clinical Center Fudan University Shanghai China
| | - Dan Yin
- Scientific Research Center Shanghai Public Health Clinical Center Fudan University Shanghai China
| | - Junyang Yang
- Department of Infectious Diseases and Immunology Shanghai Public Health Clinical Center Fudan University Shanghai China.,Wenzhou Medical University Wenzhou Zhejiang China
| | - Hongzhou Lu
- Department of Infectious Diseases and Immunology Shanghai Public Health Clinical Center Fudan University Shanghai China
| |
Collapse
|
27
|
Martínez-Flores D, Zepeda-Cervantes J, Cruz-Reséndiz A, Aguirre-Sampieri S, Sampieri A, Vaca L. SARS-CoV-2 Vaccines Based on the Spike Glycoprotein and Implications of New Viral Variants. Front Immunol 2021; 12:701501. [PMID: 34322129 PMCID: PMC8311925 DOI: 10.3389/fimmu.2021.701501] [Citation(s) in RCA: 118] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/28/2021] [Indexed: 12/12/2022] Open
Abstract
Coronavirus 19 Disease (COVID-19) originating in the province of Wuhan, China in 2019, is caused by the severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), whose infection in humans causes mild or severe clinical manifestations that mainly affect the respiratory system. So far, the COVID-19 has caused more than 2 million deaths worldwide. SARS-CoV-2 contains the Spike (S) glycoprotein on its surface, which is the main target for current vaccine development because antibodies directed against this protein can neutralize the infection. Companies and academic institutions have developed vaccines based on the S glycoprotein, as well as its antigenic domains and epitopes, which have been proven effective in generating neutralizing antibodies. However, the emergence of new SARS-CoV-2 variants could affect the effectiveness of vaccines. Here, we review the different types of vaccines designed and developed against SARS-CoV-2, placing emphasis on whether they are based on the complete S glycoprotein, its antigenic domains such as the receptor-binding domain (RBD) or short epitopes within the S glycoprotein. We also review and discuss the possible effectiveness of these vaccines against emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Daniel Martínez-Flores
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jesús Zepeda-Cervantes
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Adolfo Cruz-Reséndiz
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Sergio Aguirre-Sampieri
- Laboratorio de Fisicoquímica e Ingeniería de Proteínas, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alicia Sampieri
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Luis Vaca
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
28
|
Hodgson D, Colton H, Hornsby H, Brown R, Mckenzie J, Bradley KL, James C, Lindsey BB, Birch S, Marsh L, Wood S, Bayley M, Dickson G, James DC, Nicklin MJH, Sayers JR, Zafred D, Rowland-Jones SL, Kudesia G, Kucharski A, Darton TC, de Silva TI, Collini PJ. Risk factors for SARS-CoV-2 seroprevalence following the first pandemic wave in UK healthcare workers in a large NHS Foundation Trust. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.07.07.21260151. [PMID: 34268521 PMCID: PMC8282110 DOI: 10.1101/2021.07.07.21260151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
BACKGROUND We aimed to measure SARS-CoV-2 seroprevalence in a cohort of healthcare workers (HCWs) during the first UK wave of the COVID-19 pandemic, explore risk factors associated with infection, and investigate the impact of antibody titres on assay sensitivity. METHODS HCWs at Sheffield Teaching Hospitals NHS Foundation Trust (STH) were prospectively enrolled and sampled at two time points. SARS-CoV-2 antibodies were tested using an in-house assay for IgG and IgA reactivity against Spike and Nucleoprotein (sensitivity 99·47%, specificity 99·56%). Data were analysed using three statistical models: a seroprevalence model, an antibody kinetics model, and a heterogeneous sensitivity model. FINDINGS As of 12th June 2020, 24·4% (n=311/1275) HCWs were seropositive. Of these, 39·2% (n=122/311) were asymptomatic. The highest adjusted seroprevalence was measured in HCWs on the Acute Medical Unit (41·1%, 95% CrI 30·0-52·9) and in Physiotherapists and Occupational Therapists (39·2%, 95% CrI 24·4-56·5). Older age groups showed overall higher median antibody titres. Further modelling suggests that, for a serological assay with an overall sensitivity of 80%, antibody titres may be markedly affected by differences in age, with sensitivity estimates of 89% in those over 60 years but 61% in those ≤30 years. INTERPRETATION HCWs in acute medical units working closely with COVID-19 patients were at highest risk of infection, though whether these are infections acquired from patients or other staff is unknown. Current serological assays may underestimate seroprevalence in younger age groups if validated using sera from older and/or more symptomatic individuals. RESEARCH IN CONTEXT Evidence before this study: We searched PubMed for studies published up to March 6th 2021, using the terms "COVID", "SARS-CoV-2", "seroprevalence", and "healthcare workers", and in addition for articles of antibody titres in different age groups against coronaviruses using "coronavirus", "SARS-CoV-2, "antibody", "antibody tires", "COVID" and "age". We included studies that used serology to estimate prevalence in healthcare workers. SARS-CoV-2 seroprevalence has been shown to be greater in healthcare workers working on acute medical units or within domestic services. Antibody levels against seasonal coronaviruses, SARS-CoV and SARS-CoV-2 were found to be higher in older adults, and patients who were hospitalised.Added value of this study: In this healthcare worker seroprevalence modelling study at a large NHS foundation trust, we confirm that those working on acute medical units, COVID-19 "Red Zones" and within domestic services are most likely to be seropositive. Furthermore, we show that physiotherapists and occupational therapists have an increased risk of COVID-19 infection. We also confirm that antibody titres are greater in older individuals, even in the context of non-hospitalised cases. Importantly, we demonstrate that this can result in age-specific sensitivity in serological assays, where lower antibody titres in younger individuals results in lower assay sensitivity.Implications of all the available evidence: There are distinct occupational roles and locations in hospitals where the risk of COVID-19 infection to healthcare workers is greatest, and this knowledge should be used to prioritise infection prevention control and other measures to protect healthcare workers. Serological assays may have different sensitivity profiles across different age groups, especially if assay validation was undertaken using samples from older and/or hospitalised patients, who tend to have higher antibody titres. Future seroprevalence studies should consider adjusting for age-specific assay sensitivities to estimate true seroprevalence rates. AUTHOR CONTRIBUTIONS
Collapse
Affiliation(s)
- David Hodgson
- Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, UK
| | - Hayley Colton
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals NHS Foundation Trust, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, UK
| | - Hailey Hornsby
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, UK
| | - Rebecca Brown
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, UK
| | - Joanne Mckenzie
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, UK
| | - Kirsty L Bradley
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, UK
| | - Cameron James
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, UK
| | - Benjamin B Lindsey
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals NHS Foundation Trust, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, UK
| | - Sarah Birch
- Academic Directorate of Communicable Diseases and Specialised Medicine, Sheffield Teaching Hospitals NHS Foundation Trust, UK
| | - Louise Marsh
- Academic Directorate of Communicable Diseases and Specialised Medicine, Sheffield Teaching Hospitals NHS Foundation Trust, UK
| | - Steven Wood
- Department of Scientific Computing and Informatics, Sheffield Teaching Hospitals NHS Foundation Trust, UK
| | - Martin Bayley
- Department of Scientific Computing and Informatics, Sheffield Teaching Hospitals NHS Foundation Trust, UK
| | - Gary Dickson
- Department of Scientific Computing and Informatics, Sheffield Teaching Hospitals NHS Foundation Trust, UK
| | - David C James
- Department of Chemical and Biological Engineering, University of Sheffield, UK
| | - Martin J H Nicklin
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, UK
| | - Jon R Sayers
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, UK
- Sheffield Institute for Nucleic Acids, University of Sheffield, UK
| | - Domen Zafred
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, UK
| | - Sarah L Rowland-Jones
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals NHS Foundation Trust, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, UK
| | - Goura Kudesia
- Department of Virology, Sheffield Teaching Hospitals NHS Foundation Trust, UK
| | - Adam Kucharski
- Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, UK
| | - Thomas C Darton
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals NHS Foundation Trust, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, UK
| | - Thushan I de Silva
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals NHS Foundation Trust, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, UK
| | - Paul J Collini
- South Yorkshire Regional Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals NHS Foundation Trust, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, UK
| |
Collapse
|
29
|
Hussman JP. Severe Clinical Worsening in COVID-19 and Potential Mechanisms of Immune-Enhanced Disease. Front Med (Lausanne) 2021; 8:637642. [PMID: 34239884 PMCID: PMC8258105 DOI: 10.3389/fmed.2021.637642] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 05/31/2021] [Indexed: 12/12/2022] Open
Abstract
Infection by the novel SARS-CoV-2 coronavirus produces a range of outcomes, with the majority of cases producing mild or asymptomatic effects, and a smaller subset progressing to critical or fatal COVID-19 disease featuring severe acute respiratory distress. Although the mechanisms driving severe disease progression remain unknown, it is possible that the abrupt clinical deterioration observed in patients with critical disease corresponds to a discrete underlying expansion of viral tropism, from infection of cells comprising respiratory linings and alveolar epithelia to direct infection and activation of inflammatory monocytes and macrophages. Dysregulated immune responses could then contribute to disease severity. This article discusses the potential role of monocyte/macrophage (Mo/Mϕ) infection by SARS-CoV-2 in mediating the immune response in severe COVID-19. Additional mechanisms of immune-enhanced disease, comprising maladaptive immune responses that may aggravate rather than alleviate severity, are also discussed. Severe acute clinical worsening in COVID-19 patients may be influenced by the emergence of antibodies that participate in hyperinflammatory monocyte response, release of neutrophil extracellular traps (NETs), thrombosis, platelet apoptosis, viral entry into Fc gamma receptor (FcγR)-expressing immune cells, and induction of autoantibodies with cross-reactivity against host proteins. While the potential roles of Mo/Mϕ infection and immune-enhanced pathology in COVID-19 are consistent with a broad range of clinical and laboratory findings, their prominence remains tentative pending further validation. In the interim, these proposed mechanisms present immediate avenues of inquiry that may help to evaluate the safety of candidate vaccines and antibody-based therapeutics, and to support consideration of pathway-informed, well-tolerated therapeutic candidates targeting the dysregulated immune response.
Collapse
|
30
|
Huang CG, Dutta A, Huang CT, Chang PY, Hsiao MJ, Hsieh YC, Lin SM, Shih SR, Tsao KC, Yang CT. Relative COVID-19 Viral Persistence and Antibody Kinetics. Pathogens 2021; 10:pathogens10060752. [PMID: 34199240 PMCID: PMC8231980 DOI: 10.3390/pathogens10060752] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 12/15/2022] Open
Abstract
A total of 15 RT-PCR confirmed COVID-19 patients were admitted to our hospital during the in-itial outbreak in Taiwan. The average time of virus clearance was delayed in seven patients, 24.14 ± 4.33 days compared to 10.25 ± 0.56 days post-symptom onset (PSO) in the other eight pa-tients. There was strong antibody response in patients with viral persistence at the pharynx, with peak values of serum antibody 677.2 ± 217.8 vs. 76.70 ± 32.11 in patients with delayed versus rapid virus clearance. The patients with delayed viral clearance had excessive antibodies of compromised quality in an early stage with the delay in peak virus neutralization efficacy, 34.14 ± 7.15 versus 12.50 ± 2.35 days PSO in patients with rapid virus clearance. Weak antibody re-sponse of patients with rapid viral clearance was also effective, with substantial and comparable neutralization efficacy, 35.70 ± 8.78 versus 41.37 ± 11.49 of patients with delayed virus clearance. Human Cytokine 48-Plex Screening of the serial sera samples revealed elevated concentrations of proinflammatory cytokines and chemokines in a deceased patient with delayed virus clear-ance and severe disease. The levels were comparatively less in the other two patients who suf-fered from severe disease but eventually survived.
Collapse
Affiliation(s)
- Chung-Guei Huang
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Taoyuan 33333, Taiwan; (C.-G.H.); (P.-Y.C.); (M.-J.H.); (S.-R.S.); (K.-C.T.)
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Avijit Dutta
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Division of Infectious Diseases, Department of Medicine, Chang Gung Memorial Hospital, Taoyuan 33333, Taiwan;
| | - Ching-Tai Huang
- Division of Infectious Diseases, Department of Medicine, Chang Gung Memorial Hospital, Taoyuan 33333, Taiwan;
- Division of Infectious Diseases, Department of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Pi-Yueh Chang
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Taoyuan 33333, Taiwan; (C.-G.H.); (P.-Y.C.); (M.-J.H.); (S.-R.S.); (K.-C.T.)
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Mei-Jen Hsiao
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Taoyuan 33333, Taiwan; (C.-G.H.); (P.-Y.C.); (M.-J.H.); (S.-R.S.); (K.-C.T.)
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yu-Chia Hsieh
- Division of Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan 33333, Taiwan;
- Division of Infectious Diseases, Department of Pediatrics, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Shu-Min Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taoyuan 33333, Taiwan;
- Department of Respiratory Therapy, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Shin-Ru Shih
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Taoyuan 33333, Taiwan; (C.-G.H.); (P.-Y.C.); (M.-J.H.); (S.-R.S.); (K.-C.T.)
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Kuo-Chien Tsao
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Taoyuan 33333, Taiwan; (C.-G.H.); (P.-Y.C.); (M.-J.H.); (S.-R.S.); (K.-C.T.)
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Cheng-Ta Yang
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taoyuan 33333, Taiwan;
- Department of Respiratory Therapy, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Correspondence: ; Tel.: +886-3-3281200
| |
Collapse
|
31
|
Zhao Z, Huang C, Huang Z, Lin F, He Q, Tao D, Jaffrezic-Renault N, Guo Z. Advancements in electrochemical biosensing for respiratory virus detection: A review. Trends Analyt Chem 2021; 139:116253. [PMID: 33727755 PMCID: PMC7952277 DOI: 10.1016/j.trac.2021.116253] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Respiratory viruses are real menace for human health which result in devastating epidemic disease. Consequently, it is in urgent need of identifying and quantifying virus with a rapid, sensitive and precise approach. The study of electrochemical biosensors for respiratory virus detection has become one of the most rapidly developing scientific fields. Recent developments in electrochemical biosensors concerning respiratory virus detection are comprehensively reviewed in this paper. This review is structured along common detecting objects of respiratory viruses, electrochemical biosensors, electrochemical biosensors for respiratory virus detection and future challenges. The electrochemical biosensors for respiratory virus detection are introduced, including nucleic acids-based, immunosensors and other affinity biosensors. Lastly, for Coronavirus disease 2019 (COVID-19) diagnosis, the future challenges regarding developing electrochemical biosensor-based Point-of-Care Tests (POCTs) are summarized. This review is expected to provide a helpful guide for the researchers entering this interdisciplinary field and developing more novel electrochemical biosensors for respiratory virus detection.
Collapse
Affiliation(s)
- Zhi Zhao
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan 430065, PR China
- School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan 430065, PR China
| | - Changfu Huang
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan 430065, PR China
- School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan 430065, PR China
| | - Ziyu Huang
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan 430065, PR China
- School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan 430065, PR China
| | - Fengjuan Lin
- School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan 430065, PR China
| | - Qinlin He
- School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan 430065, PR China
| | - Dan Tao
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan 430065, PR China
- School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan 430065, PR China
| | - Nicole Jaffrezic-Renault
- University of Lyon, Institute of Analytical Sciences, UMR-CNRS 5280, 5, La Doua Street, Villeurbanne 69100, France
| | - Zhenzhong Guo
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan 430065, PR China
| |
Collapse
|
32
|
Taylor PC, Adams AC, Hufford MM, de la Torre I, Winthrop K, Gottlieb RL. Neutralizing monoclonal antibodies for treatment of COVID-19. Nat Rev Immunol 2021; 21:382-393. [PMID: 33875867 PMCID: PMC8054133 DOI: 10.1038/s41577-021-00542-x] [Citation(s) in RCA: 486] [Impact Index Per Article: 162.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2021] [Indexed: 12/15/2022]
Abstract
Several neutralizing monoclonal antibodies (mAbs) to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been developed and are now under evaluation in clinical trials. With the US Food and Drug Administration recently granting emergency use authorizations for neutralizing mAbs in non-hospitalized patients with mild-to-moderate COVID-19, there is an urgent need to discuss the broader potential of these novel therapies and to develop strategies to deploy them effectively in clinical practice, given limited initial availability. Here, we review the precedent for passive immunization and lessons learned from using antibody therapies for viral infections such as respiratory syncytial virus, Ebola virus and SARS-CoV infections. We then focus on the deployment of convalescent plasma and neutralizing mAbs for treatment of SARS-CoV-2. We review specific clinical questions, including the rationale for stratification of patients, potential biomarkers, known risk factors and temporal considerations for optimal clinical use. To answer these questions, there is a need to understand factors such as the kinetics of viral load and its correlation with clinical outcomes, endogenous antibody responses, pharmacokinetic properties of neutralizing mAbs and the potential benefit of combining antibodies to defend against emerging viral variants.
Collapse
MESH Headings
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Neutralizing/therapeutic use
- Antibodies, Viral/therapeutic use
- Antibody-Dependent Enhancement
- COVID-19/immunology
- COVID-19/therapy
- COVID-19/virology
- Drug Development
- Drug Resistance, Viral/genetics
- Drug Resistance, Viral/immunology
- Humans
- Immunization, Passive/adverse effects
- Immunization, Passive/methods
- Models, Immunological
- Pandemics
- SARS-CoV-2/drug effects
- SARS-CoV-2/genetics
- SARS-CoV-2/immunology
- COVID-19 Serotherapy
Collapse
Affiliation(s)
- Peter C Taylor
- Botnar Research Centre, University of Oxford, Oxford, UK.
| | | | | | | | | | - Robert L Gottlieb
- Baylor University Medical Center, Dallas, TX, USA
- Baylor Scott & White Research Institute, Dallas, TX, USA
| |
Collapse
|
33
|
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: 23.3] [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
|
34
|
Hasan A, Al-Ozairi E, Al-Baqsumi Z, Ahmad R, Al-Mulla F. Cellular and Humoral Immune Responses in Covid-19 and Immunotherapeutic Approaches. Immunotargets Ther 2021; 10:63-85. [PMID: 33728277 PMCID: PMC7955763 DOI: 10.2147/itt.s280706] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 12/22/2020] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (Covid-19), caused by the novel coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), can range in severity from asymptomatic to severe/critical disease. SARS-CoV-2 uses angiotensin-converting enzyme 2 to infect cells leading to a strong inflammatory response, which is most profound in patients who progress to severe Covid-19. Recent studies have begun to unravel some of the differences in the innate and adaptive immune response to SARS-CoV-2 in patients with different degrees of disease severity. These studies have attributed the severe form of Covid-19 to a dysfunctional innate immune response, such as a delayed and/or deficient type I interferon response, coupled with an exaggerated and/or a dysfunctional adaptive immunity. Differences in T-cell (including CD4+ T-cells, CD8+ T-cells, T follicular helper cells, γδ-T-cells, and regulatory T-cells) and B-cell (transitional cells, double-negative 2 cells, antibody-secreting cells) responses have been identified in patients with severe disease compared to mild cases. Moreover, differences in the kinetic/titer of neutralizing antibody responses have been described in severe disease, which may be confounded by antibody-dependent enhancement. Importantly, the presence of preexisting autoantibodies against type I interferon has been described as a major cause of severe/critical disease. Additionally, priorVaccine and multiple vaccine exposure, trained innate immunity, cross-reactive immunity, and serological immune imprinting may all contribute towards disease severity and outcome. Several therapeutic and preventative approaches have been under intense investigations; these include vaccines (three of which have passed Phase 3 clinical trials), therapeutic antibodies, and immunosuppressants.
Collapse
Affiliation(s)
- Amal Hasan
- Department of Immunology and Microbiology, Research Division, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| | - Ebaa Al-Ozairi
- Clinical Research Unit, Medical Division, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
- Department of Medicine, Faculty of Medicine, Jabriya, Kuwait City, Kuwait
| | - Zahraa Al-Baqsumi
- Department of Immunology and Microbiology, Research Division, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| | - Rasheed Ahmad
- Department of Immunology and Microbiology, Research Division, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| | - Fahd Al-Mulla
- Department of Genetics and Bioinformatics, Functional Genomics, Research Division, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| |
Collapse
|
35
|
Jansen van Vuren E, Steyn SF, Brink CB, Möller M, Viljoen FP, Harvey BH. The neuropsychiatric manifestations of COVID-19: Interactions with psychiatric illness and pharmacological treatment. Biomed Pharmacother 2021; 135:111200. [PMID: 33421734 PMCID: PMC7834135 DOI: 10.1016/j.biopha.2020.111200] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/15/2020] [Accepted: 12/26/2020] [Indexed: 12/12/2022] Open
Abstract
The recent outbreak of the corona virus disease (COVID-19) has had major global impact. The relationship between severe acute respiratory syndrome coronavirus (SARS-CoV-2) infection and psychiatric diseases is of great concern, with an evident link between corona virus infections and various central and peripheral nervous system manifestations. Unmitigated neuro-inflammation has been noted to underlie not only the severe respiratory complications of the disease but is also present in a range of neuro-psychiatric illnesses. Several neurological and psychiatric disorders are characterized by immune-inflammatory states, while treatments for these disorders have distinct anti-inflammatory properties and effects. With inflammation being a common contributing factor in SARS-CoV-2, as well as psychiatric disorders, treatment of either condition may affect disease progression of the other or alter response to pharmacological treatment. In this review, we elucidate how viral infections could affect pre-existing psychiatric conditions and how pharmacological treatments of these conditions may affect overall progress and outcome in the treatment of SARS-CoV-2. We address whether any treatment-induced benefits and potential adverse effects may ultimately affect the overall treatment approach, considering the underlying dysregulated neuro-inflammatory processes and potential drug interactions. Finally, we suggest adjunctive treatment options for SARS-CoV-2-associated neuro-psychiatric symptoms.
Collapse
Affiliation(s)
- Esmé Jansen van Vuren
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa.
| | - Stephan F Steyn
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Christiaan B Brink
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Marisa Möller
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Francois P Viljoen
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Brian H Harvey
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa; South African MRC Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
36
|
Folgueira MD, Luczkowiak J, Lasala F, Pérez-Rivilla A, Delgado R. Prolonged SARS-CoV-2 cell culture replication in respiratory samples from patients with severe COVID-19. Clin Microbiol Infect 2021; 27:886-891. [PMID: 33631334 PMCID: PMC7898982 DOI: 10.1016/j.cmi.2021.02.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/06/2021] [Accepted: 02/10/2021] [Indexed: 01/13/2023]
Abstract
Objectives This study compares the infectivity of SARS-CoV-2 in respiratory samples from patients with mild COVID-19 with those from hospitalized patients with severe bilateral pneumonia. In severe COVID-19, we also analysed the presence of neutralizing activity in paired sera. Methods We performed cell cultures on 193 real-time reverse transcription polymerase chain reaction respiratory samples, positive for SARS-CoV-2, obtained from 189 patients at various times, from clinical diagnosis to follow-up. Eleven samples were obtained from asymptomatic individuals, 91 samples from 91 outpatients with mild forms of COVID-19 and 91 samples from 87 inpatients with severe pneumonia. In these patients, neutralizing activity was analysed in 30 paired sera collected after symptom onset >10 days. Results We detected a cytopathic effect (CPE) in 91/193 (47%) samples. Viral viability was maintained for up to 10 days in patients with mild COVID-19. In patients with severe COVID-19, the virus remained viable for up to 32 days after the onset of symptoms. Patients with severe COVID-19 presented infectious virus at a significantly higher rate in the samples with moderate to low viral load (cycle threshold value ≥ 26): 32/75 (43%) versus 14/63 (22%) for mild cases (p < 0.01). We observed a positive CPE despite the presence of clear neutralizing activity (NT50 > 1:1024 in 10% (3/30) of samples. Discussion Patients with severe COVID-19 might shed viable virus during prolonged periods of up to 4 weeks after symptom onset, even when presenting high cycle threshold values in their respiratory samples and despite having developed high neutralizing antibody titres.
Collapse
Affiliation(s)
- Maria Dolores Folgueira
- Department of Clinical Microbiology, Hospital Universitario 12 de Octubre, Madrid, Spain; Research Institute Hospital 12 de Octubre, imas12, Madrid, Spain; Department of Medicine, Complutense University of Madrid, School of Medicine, Madrid, Spain.
| | | | - Fátima Lasala
- Research Institute Hospital 12 de Octubre, imas12, Madrid, Spain
| | - Alfredo Pérez-Rivilla
- Department of Clinical Microbiology, Hospital Universitario 12 de Octubre, Madrid, Spain; Research Institute Hospital 12 de Octubre, imas12, Madrid, Spain
| | - Rafael Delgado
- Department of Clinical Microbiology, Hospital Universitario 12 de Octubre, Madrid, Spain; Research Institute Hospital 12 de Octubre, imas12, Madrid, Spain; Department of Medicine, Complutense University of Madrid, School of Medicine, Madrid, Spain
| |
Collapse
|
37
|
Exploring beyond clinical routine SARS-CoV-2 serology using MultiCoV-Ab to evaluate endemic coronavirus cross-reactivity. Nat Commun 2021; 12:1152. [PMID: 33608538 PMCID: PMC7896075 DOI: 10.1038/s41467-021-20973-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 01/06/2021] [Indexed: 12/11/2022] Open
Abstract
The humoral immune response to SARS-CoV-2 is a benchmark for immunity and detailed analysis is required to understand the manifestation and progression of COVID-19, monitor seroconversion within the general population, and support vaccine development. The majority of currently available commercial serological assays only quantify the SARS-CoV-2 antibody response against individual antigens, limiting our understanding of the immune response. To overcome this, we have developed a multiplex immunoassay (MultiCoV-Ab) including spike and nucleocapsid proteins of SARS-CoV-2 and the endemic human coronaviruses. Compared to three broadly used commercial in vitro diagnostic tests, our MultiCoV-Ab achieves a higher sensitivity and specificity when analyzing a well-characterized sample set of SARS-CoV-2 infected and uninfected individuals. We find a high response against endemic coronaviruses in our sample set, but no consistent cross-reactive IgG response patterns against SARS-CoV-2. Here we show a robust, high-content-enabled, antigen-saving multiplex assay suited to both monitoring vaccination studies and facilitating epidemiologic screenings for humoral immunity towards pandemic and endemic coronaviruses.
Collapse
|
38
|
Jonsdottir HR, Bielecki M, Siegrist D, Buehrer TW, Züst R, Deuel JW. Titers of Neutralizing Antibodies against SARS-CoV-2 Are Independent of Symptoms of Non-Severe COVID-19 in Young Adults. Viruses 2021; 13:v13020284. [PMID: 33673067 PMCID: PMC7918933 DOI: 10.3390/v13020284] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/08/2021] [Accepted: 02/08/2021] [Indexed: 12/20/2022] Open
Abstract
Neutralizing antibodies are an important part of the humoral immune response to SARS-CoV-2. It is currently unclear to what extent such antibodies are produced after non-severe disease or asymptomatic infection. We studied a cluster of SARS-CoV-2 infections among a homogeneous population of 332 predominantly male Swiss soldiers and determined the neutralizing antibody response with a serum neutralization assay using a recombinant SARS-CoV-2-GFP. All patients with non-severe COVID-19 showed a swift humoral response within two weeks after the onset of symptoms, which remained stable for the duration of the study. One month after the outbreak, titers in COVID-19 convalescents did not differ from the titers of asymptomatically infected individuals. Furthermore, symptoms of COVID-19 did not correlate with neutralizing antibody titers. Therefore, we conclude that asymptomatic infection can induce the same humoral immunity as non-severe COVID-19 in young adults.
Collapse
Affiliation(s)
| | - Michel Bielecki
- Swiss Armed Forces, Medical Services, Worblentalstrasse, 3063 Ittigen, Switzerland; (M.B.); (T.W.B.)
- Epidemiology, Biostatistics and Prevention Institute, Travel Clinic, Hirschengraben, University of Zurich, 8006 Zürich, Switzerland
| | - Denise Siegrist
- SPIEZ Laboratory, Austrasse, 3700 Spiez, Switzerland; (H.R.J.); (D.S.)
| | - Thomas W. Buehrer
- Swiss Armed Forces, Medical Services, Worblentalstrasse, 3063 Ittigen, Switzerland; (M.B.); (T.W.B.)
| | - Roland Züst
- SPIEZ Laboratory, Austrasse, 3700 Spiez, Switzerland; (H.R.J.); (D.S.)
- Correspondence: (R.Z.); (J.W.D.)
| | - Jeremy W. Deuel
- Swiss Armed Forces, Medical Services, Worblentalstrasse, 3063 Ittigen, Switzerland; (M.B.); (T.W.B.)
- Jeffrey Cheah Biomedical Centre, Department of Haematology and MRC—Wellcome Stem Cell Institute, Puddicombe Way, University of Cambridge, Cambridge CB2 AW, UK
- Correspondence: (R.Z.); (J.W.D.)
| |
Collapse
|
39
|
Crawford KHD, Dingens AS, Eguia R, Wolf CR, Wilcox N, Logue JK, Shuey K, Casto AM, Fiala B, Wrenn S, Pettie D, King NP, Greninger AL, Chu HY, Bloom JD. Dynamics of Neutralizing Antibody Titers in the Months After Severe Acute Respiratory Syndrome Coronavirus 2 Infection. J Infect Dis 2021; 223:197-205. [PMID: 33535236 PMCID: PMC7543487 DOI: 10.1093/infdis/jiaa618] [Citation(s) in RCA: 178] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 09/28/2020] [Indexed: 02/07/2023] Open
Abstract
Most individuals infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) develop neutralizing antibodies that target the viral spike protein. In this study, we quantified how levels of these antibodies change in the months after SARS-CoV-2 infection by examining longitudinal samples collected approximately 30-152 days after symptom onset from a prospective cohort of 32 recovered individuals with asymptomatic, mild, or moderate-severe disease. Neutralizing antibody titers declined an average of about 4-fold from 1 to 4 months after symptom onset. This decline in neutralizing antibody titers was accompanied by a decline in total antibodies capable of binding the viral spike protein or its receptor-binding domain. Importantly, our data are consistent with the expected early immune response to viral infection, where an initial peak in antibody levels is followed by a decline to a lower plateau. Additional studies of long-lived B cells and antibody titers over longer time frames are necessary to determine the durability of immunity to SARS-CoV-2.
Collapse
Affiliation(s)
- Katharine H D Crawford
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
- Medical Scientist Training Program, University of Washington, Seattle, Washington, USA
| | - Adam S Dingens
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Rachel Eguia
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Caitlin R Wolf
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Naomi Wilcox
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Jennifer K Logue
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Kiel Shuey
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Amanda M Casto
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Brooke Fiala
- Department of Biochemistry, University of Washington, Seattle, Washington, USA
- Institute for Protein Design, University of Washington, Seattle, Washington, USA
| | - Samuel Wrenn
- Department of Biochemistry, University of Washington, Seattle, Washington, USA
- Institute for Protein Design, University of Washington, Seattle, Washington, USA
| | - Deleah Pettie
- Department of Biochemistry, University of Washington, Seattle, Washington, USA
- Institute for Protein Design, University of Washington, Seattle, Washington, USA
| | - Neil P King
- Department of Biochemistry, University of Washington, Seattle, Washington, USA
- Institute for Protein Design, University of Washington, Seattle, Washington, USA
| | - Alexander L Greninger
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Helen Y Chu
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Jesse D Bloom
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
- Howard Hughes Medical Institute, Seattle, Washington, USA
| |
Collapse
|
40
|
Carrillo J, Izquierdo-Useros N, Ávila-Nieto C, Pradenas E, Clotet B, Blanco J. Humoral immune responses and neutralizing antibodies against SARS-CoV-2; implications in pathogenesis and protective immunity. Biochem Biophys Res Commun 2021; 538:187-191. [PMID: 33187644 PMCID: PMC7648524 DOI: 10.1016/j.bbrc.2020.10.108] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022]
Abstract
The magnitude and the quality of humoral responses against SARS-CoV-2 have been associated with clinical outcome. Although the elicitation of humoral responses against different viral proteins is rapid and occurs in most infected individuals, its magnitude is highly variable among them and positively correlates with COVID-19 disease severity. This rapid response is characterized by the almost concomitant appearance of virus-specific IgG, IgA and IgM antibodies that contain neutralizing antibodies directed against different epitopes of the Spike glycoprotein. Of particularly interest, the antibodies against domain of the Spike that interacts with the cellular receptor ACE2, known as the receptor binding domain (RBD), are present in most infected individuals and are block viral entry and infectivity. Such neutralizing antibodies protect different animal species when administered before virus exposure; therefore, its elicitation is the main target of current vaccine approaches and their clinical use as recombinant monoclonal antibodies (mAbs) is being explored. Yet, little information exists on the duration of humoral responses during natural infection. This is a key issue that will impact the management of the pandemic and determine the utility of seroconversion studies and the level of herd immunity. Certainly, several cases of reinfection have been reported, suggesting that immunity could be transient, as reported for other coronaviruses. In summary, although the kinetics of the generation of antibodies against SASR-CoV-2 and their protective activity have been clearly defined, their role in COVID-19 pathogenesis and the length of these responses are still open questions.
Collapse
Affiliation(s)
- Jorge Carrillo
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain,Corresponding author. Institut de Recerca de la Sida, IrsiCaixa Hospital Universitari Germans Trias i Pujol, Ctra. de Canyet s/n. 2a Planta Maternal, 08916, Badalona, Barcelona, Spain
| | - Nuria Izquierdo-Useros
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | - Carlos Ávila-Nieto
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | - Edwards Pradenas
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain,Infectious Diseases Department, Germans Trias i Pujol Hospital, Badalona, Catalonia, Spain,University of Vic (UVic-UCC), Vic, Catalonia, Spain
| | - Julià Blanco
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain,University of Vic (UVic-UCC), Vic, Catalonia, Spain,Corresponding author. Institut de Recerca de la Sida, IrsiCaixa Hospital Universitari Germans Trias i Pujol, Ctra. de Canyet s/n. 2a Planta Maternal, 08916, Badalona, Barcelona, Spain
| |
Collapse
|
41
|
Trinité B, Tarrés-Freixas F, Rodon J, Pradenas E, Urrea V, Marfil S, Rodríguez de la Concepción ML, Ávila-Nieto C, Aguilar-Gurrieri C, Barajas A, Ortiz R, Paredes R, Mateu L, Valencia A, Guallar V, Ruiz L, Grau E, Massanella M, Puig J, Chamorro A, Izquierdo-Useros N, Segalés J, Clotet B, Carrillo J, Vergara-Alert J, Blanco J. SARS-CoV-2 infection elicits a rapid neutralizing antibody response that correlates with disease severity. Sci Rep 2021; 11:2608. [PMID: 33510275 PMCID: PMC7843981 DOI: 10.1038/s41598-021-81862-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 01/08/2021] [Indexed: 12/14/2022] Open
Abstract
The protective effect of neutralizing antibodies in SARS-CoV-2 infected individuals is not yet well defined. To address this issue, we have analyzed the kinetics of neutralizing antibody responses and their association with disease severity. Between March and May 2020, the prospective KING study enrolled 72 COVID-19+ participants grouped according to disease severity. SARS-CoV-2 infection was diagnosed by serological and virological tests. Plasma neutralizing responses were assessed against replicative virus and pseudoviral particles. Multiple regression and non-parametric tests were used to analyze dependence of parameters. The magnitude of neutralizing titers significantly increased with disease severity. Hospitalized individuals developed higher titers compared to mild-symptomatic and asymptomatic individuals, which together showed titers below the detection limit in 50% of cases. Longitudinal analysis confirmed the strong differences in neutralizing titers between non-hospitalized and hospitalized participants and showed rapid kinetics of appearance of neutralizing antibodies (50% and 80% of maximal activity reached after 11 and 17 days after symptoms onset, respectively) in hospitalized patients. No significant impact of age, gender or treatment on the neutralizing titers was observed in this limited cohort. These data identify a clear association of humoral immunity with disease severity and point to immune mechanisms other than antibodies as relevant players in COVID-19 protection.
Collapse
Affiliation(s)
- Benjamin Trinité
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Ferran Tarrés-Freixas
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, 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
| | - Edwards Pradenas
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Víctor Urrea
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Silvia Marfil
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - María Luisa Rodríguez de la Concepción
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Carlos Ávila-Nieto
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Carmen Aguilar-Gurrieri
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Ana Barajas
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Raquel Ortiz
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Roger Paredes
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, 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, Badalona, Catalonia, Spain
| | - Lourdes Mateu
- Infectious Diseases Department, Fight Against AIDS Foundation (FLS), Germans Trias I Pujol Hospital, Badalona, Catalonia, Spain
| | | | - Víctor Guallar
- Barcelona Supercomputing Center, Barcelona, Catalonia, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Catalonia, Spain
| | - Lidia Ruiz
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Eulàlia Grau
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Marta Massanella
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Jordi Puig
- Infectious Diseases Department, Fight Against AIDS Foundation (FLS), Germans Trias I Pujol Hospital, Badalona, Catalonia, Spain
| | - Anna Chamorro
- Infectious Diseases Department, Fight Against AIDS Foundation (FLS), Germans Trias I Pujol Hospital, Badalona, Catalonia, Spain
| | - Nuria Izquierdo-Useros
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Joaquim Segalés
- IRTA Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la UAB, 08193, Bellaterra, Catalonia, Spain.,UAB, CReSA (IRTA-UAB), Campus de la UAB, 08193, Bellaterra, Cerdanyola del Vallès, Catalonia, Spain
| | - Bonaventura Clotet
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, 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, Badalona, Catalonia, Spain.,University of Vic-Central University of Catalonia (UVic-UCC), Vic, Catalonia, Spain
| | - Jorge Carrillo
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Júlia Vergara-Alert
- IRTA Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la UAB, 08193, Bellaterra, Catalonia, Spain
| | - Julià Blanco
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain. .,University of Vic-Central University of Catalonia (UVic-UCC), Vic, Catalonia, Spain.
| |
Collapse
|
42
|
Rothan HA, Byrareddy SN. The potential threat of multisystem inflammatory syndrome in children during the COVID-19 pandemic. Pediatr Allergy Immunol 2021; 32:17-22. [PMID: 32897642 PMCID: PMC7887110 DOI: 10.1111/pai.13361] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 08/15/2020] [Accepted: 08/26/2020] [Indexed: 12/16/2022]
Abstract
Multisystem inflammatory syndrome in children (MIS-C) during the COVID-19 pandemic raised a global alert from the Centers for Disease Control and Prevention's Health Alert Network. The main manifestations of MIS-C (also known as pediatric MIS (PMIS)) in the setting of a severe inflammatory state include fever, diarrhea, shock, and variable presence of rash, conjunctivitis, extremity edema, and mucous membrane changes. In some cases, these symptoms progressed to multi-organ failure. The low percentage of children with asymptomatic cases compared with mild illness and moderate illness could be correlated with the rare cases of MIS-C. One potential explanation for the progression to severe MIS-C disease despite the presence of readily detectable anti-SARS-CoV-2 antibodies could be due to the potential role of antibody-dependent enhancement (ADE). We reason that the incidence of the ADE phenomenon whereby the pathogen-specific antibodies can promote pathology should be considered in vaccine development against SARS-CoV-2.
Collapse
Affiliation(s)
- Hussin A. Rothan
- Department of Biology, College of Arts and Sciences, Georgia State University, GA, USA
| | - Siddappa N. Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Centre, Omaha, NE, USA
- Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Centre, Omaha, NE, USA
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Centre, Omaha, NE, USA
| |
Collapse
|
43
|
Xu L, Ma Z, Li Y, Pang Z, Xiao S. Antibody dependent enhancement: Unavoidable problems in vaccine development. Adv Immunol 2021; 151:99-133. [PMID: 34656289 PMCID: PMC8438590 DOI: 10.1016/bs.ai.2021.08.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In some cases, antibodies can enhance virus entry and replication in cells. This phenomenon is called antibody-dependent infection enhancement (ADE). ADE not only promotes the virus to be recognized by the target cell and enters the target cell, but also affects the signal transmission in the target cell. Early formalin-inactivated virus vaccines such as aluminum adjuvants (RSV and measles) have been shown to induce ADE. Although there is no direct evidence that there is ADE in COVID-19, this potential risk is a huge challenge for prevention and vaccine development. This article focuses on the virus-induced ADE phenomenon and its molecular mechanism. It also summarizes various attempts in vaccine research and development to eliminate the ADE phenomenon, and proposes to avoid ADE in vaccine development from the perspective of antigens and adjuvants.
Collapse
|
44
|
Bobik TV, Kostin NN, Skryabin GA, Tsabai PN, Simonova MA, Knorre VD, Stratienko ON, Aleshenko NL, Vorobiev II, Khurs EN, Mokrushina YA, Smirnov IV, Alekhin AI, Nikitin AE, Gabibov AG. COVID-19 in Russia: Clinical and Immunological Features of the First-Wave Patients. Acta Naturae 2021; 13:102-115. [PMID: 33959390 PMCID: PMC8084292 DOI: 10.32607/actanaturae.11374] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 02/16/2021] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease outbreak in 2019 (COVID-19) has now achieved the level of a global pandemic and affected more than 100 million people on all five continents and caused over 2 million deaths. Russia is, needless to say, among the countries affected by SARS-CoV-2, and its health authorities have mobilized significant efforts and resources to fight the disease. The paper presents the result of a functional analysis of 155 patients in the Moscow Region who were examined at the Central Clinical Hospital of the Russian Academy of Sciences during the first wave of the pandemic (February-July, 2020). The inclusion criteria were a positive PCR test and typical, computed tomographic findings of viral pneumonia in the form of ground-glass opacities. A clinical correlation analysis was performed in four groups of patients: (1) those who were not on mechanical ventilation, (2) those who were on mechanical ventilation, and (3) those who subsequently recovered or (4) died. The correlation analysis also considered confounding comorbidities (diabetes, metabolic syndrome, hypertension, etc.). The immunological status of the patients was examined (levels of immunoglobulins of the M, A, G classes and their subclasses, as well as the total immunoglobulin level) using an original SARS-CoV-2 antibody ELISA kit. The ELISA kit was developed using linear S-protein RBD-SD1 and NTD fragments, as well as the N-protein, as antigens. These antigens were produced in the prokaryotic E. coli system. Recombinant RBD produced in the eukaryotic CHO system (RBD CHO) was used as an antigen representing conformational RBD epitopes. The immunoglobulin A level was found to be the earliest serological criterion for the development of a SARS-CoV-2 infection and it yielded the best sensitivity and diagnostic significance of ELISA compared to that of class M immunoglobulin. We demonstrated that the seroconversion rate of "early" N-protein-specific IgM and IgA antibodies is comparable to that of antibodies specific to RBD conformational epitopes. At the same time, seroconversion of SARS-CoV-2 N-protein-specific class G immunoglobulins was significantly faster compared to that of other specific antibodies. Our findings suggest that the strong immunogenicity of the RBD fragment is for the most part associated with its conformational epitopes, while the linear RBD and NTD epitopes have the least immunogenicity. An analysis of the occurrence rate of SARS-CoV-2-specific immunoglobulins of different classes revealed that RBD- and N-specific antibodies should be evaluated in parallel to improve the sensitivity of ELISA. An analysis of the immunoglobulin subclass distribution in sera of seropositive patients revealed uniform induction of N-protein-specific IgG subclasses G1-G4 and IgA subclasses A1-A2 in groups of patients with varying severity of COVID-19. In the case of the S-protein, G1, G3, and A1 were the main subclasses of antibodies involved in the immune response.
Collapse
Affiliation(s)
- T. V. Bobik
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997 Russia
| | - N. N. Kostin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997 Russia
| | - G. A. Skryabin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997 Russia
| | - P. N. Tsabai
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997 Russia
| | - M. A. Simonova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997 Russia
| | - V. D. Knorre
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997 Russia
| | - O. N. Stratienko
- Central Clinical Hospital of the Russian Academy of Sciences, Moscow, 117593 Russia
| | - N. L. Aleshenko
- Central Clinical Hospital of the Russian Academy of Sciences, Moscow, 117593 Russia
| | - I. I. Vorobiev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997 Russia
| | - E. N. Khurs
- Engelhardt Institute of Molecular Biology, RAS, Moscow, 119991 Russia
| | - Yu. A. Mokrushina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997 Russia
| | - I. V. Smirnov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997 Russia
| | - A. I. Alekhin
- Central Clinical Hospital of the Russian Academy of Sciences, Moscow, 117593 Russia
| | - A. E. Nikitin
- Central Clinical Hospital of the Russian Academy of Sciences, Moscow, 117593 Russia
| | - A. G. Gabibov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997 Russia
| |
Collapse
|
45
|
Abstract
The newly emerged severe acute respiratory syndrome-related coronavirus-2 (SARS-CoV-2) has recently caused pandemic Coronavirus Disease-2019 (COVID-19). Considering the serious medical, economic and social consequences of this pandemic and the lack of definite medication and vaccine it is necessary to describe natural immune responses to the SARS-CoV-2 in order to exploit them for treating the patients and monitoring the general population. Moreover, detecting the most immunogenic antigens of the virus is fundamental for designing effective vaccines. Antibodies being valuable for diagnostic therapeutic and protective purposes are suitable to be addressed in this context. Herein, we have summarized the findings of serological investigations and the outcomes of neutralizing antibodies administration in COVID-19 patients.
Collapse
Affiliation(s)
- Sara Assadiasl
- Molecular Immunology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Yousef Fatahi
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Zavvar
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Nicknam
- Molecular Immunology Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
46
|
Lucas C, Klein J, Sundaram M, Liu F, Wong P, Silva J, Mao T, Oh JE, Tokuyama M, Lu P, Venkataraman A, Park A, Israelow B, Wyllie AL, Vogels CBF, Muenker MC, Casanovas-Massana A, Schulz WL, Zell J, Campbell M, Fournier JB, Grubaugh ND, Farhadian S, Wisnewski AV, Cruz CD, Omer S, Ko AI, Ring A, Iwasaki A. Kinetics of antibody responses dictate COVID-19 outcome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.12.18.20248331. [PMID: 33398304 DOI: 10.1101/2020.06.13.20130252] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Recent studies have provided insights into innate and adaptive immune dynamics in coronavirus disease 2019 (COVID-19). Yet, the exact feature of antibody responses that governs COVID-19 disease outcomes remain unclear. Here, we analysed humoral immune responses in 209 asymptomatic, mild, moderate and severe COVID-19 patients over time to probe the nature of antibody responses in disease severity and mortality. We observed a correlation between anti-Spike (S) IgG levels, length of hospitalization and clinical parameters associated with worse clinical progression. While high anti-S IgG levels correlated with worse disease severity, such correlation was time-dependent. Deceased patients did not have higher overall humoral response than live discharged patients. However, they mounted a robust, yet delayed response, measured by anti-S, anti-RBD IgG, and neutralizing antibody (NAb) levels, compared to survivors. Delayed seroconversion kinetics correlated with impaired viral control in deceased patients. Finally, while sera from 89% of patients displayed some neutralization capacity during their disease course, NAb generation prior to 14 days of disease onset emerged as a key factor for recovery. These data indicate that COVID-19 mortality does not correlate with the cross-sectional antiviral antibody levels per se , but rather with the delayed kinetics of NAb production.
Collapse
|
47
|
Ren L, Zhang L, Chang D, Wang J, Hu Y, Chen H, Guo L, Wu C, Wang C, Wang Y, Wang Y, Wang G, Yang S, Dela Cruz CS, Sharma L, Wang L, Zhang D, Wang J. The kinetics of humoral response and its relationship with the disease severity in COVID-19. Commun Biol 2020; 3:780. [PMID: 33311543 PMCID: PMC7733479 DOI: 10.1038/s42003-020-01526-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
Coronavirus Disease 2019 (COVID-19) has caused a global pandemic. Here we profiled the humoral response against Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) by measuring immunoglobulin (Ig) A, IgM, and IgG against nucleocapsid and spike proteins, along with IgM and IgG antibodies against receptor-binding domain (RBD) of the spike protein and total neutralizing antibodies (NAbs). We tested 279 plasma samples collected from 176 COVID-19 patients who presented and enrolled at different stages of their disease. Plasma dilutions were optimized and based on the data, a single dilution of plasma was used. The mean absorbance at 450 nm was measured for Ig levels and NAbs were measured using geometric mean titers. We demonstrate that more severe cases have a late-onset in the humoral response compared to mild/moderate infections. All the antibody titers continue to rise in patients with COVID-19 over the disease course. However, these levels are mostly unrelated to disease severity. The appearance time and titers of NAbs showed a significant positive correlation to the antibodies against spike protein. Our results suggest the late onset of antibody response as a risk factor for disease severity, however, there is a limited role of antibody titers in predicting disease severity of COVID-19.
Collapse
Affiliation(s)
- Lili Ren
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, P.R. China
- Key Laboratory of Respiratory Disease Pathogenomics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China
| | - Lulu Zhang
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, P.R. China
- Key Laboratory of Respiratory Disease Pathogenomics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China
| | - De Chang
- Third Medical Center of Chinese PLA General Hospital, Beijing, 100039, P.R. China
| | - Junwen Wang
- Wuhan 4th hospital, Wuhan, 430023, P.R. China
| | - Yongfeng Hu
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, P.R. China
| | - Hong Chen
- The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, P.R. China
| | - Li Guo
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, P.R. China
- Key Laboratory of Respiratory Disease Pathogenomics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China
| | - Chao Wu
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, P.R. China
| | - Conghui Wang
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, P.R. China
| | - Yingying Wang
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, P.R. China
| | - Ying Wang
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, P.R. China
| | - Geng Wang
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, P.R. China
| | - Siyuan Yang
- Laboratory of Infectious Diseases Center of Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, P.R. China
| | - Charles S Dela Cruz
- Section of Pulmonary and Critical Care and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Lokesh Sharma
- Section of Pulmonary and Critical Care and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.
| | - Linghang Wang
- Emergency Department of Infectious Diseases of Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, P.R. China.
| | - Dingyu Zhang
- Research Center for Translational Medicine, Wuhan Jin Yin-tan Hospital, Wuhan, 430023, P.R. China.
- Wuhan Research Center for Infectious Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Wuhan, 430023, P.R. China.
| | - Jianwei Wang
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, P.R. China.
- Key Laboratory of Respiratory Disease Pathogenomics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China.
| |
Collapse
|
48
|
Kim DS, Rowland-Jones S, Gea-Mallorquí E. Will SARS-CoV-2 Infection Elicit Long-Lasting Protective or Sterilising Immunity? Implications for Vaccine Strategies (2020). Front Immunol 2020; 11:571481. [PMID: 33362759 PMCID: PMC7756008 DOI: 10.3389/fimmu.2020.571481] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 11/06/2020] [Indexed: 12/20/2022] Open
Abstract
In December 2019, an outbreak of a novel coronavirus (SARS-CoV-2) in Wuhan, China resulted in the current COVID-19 global pandemic. The human immune system has not previously encountered this virus, raising the important question as to whether or not protective immunity is generated by infection. Growing evidence suggests that protective immunity can indeed be acquired post-infection-although a handful of reinfection cases have been reported. However, it is still unknown whether the immune response to SARS-CoV-2 leads to some degree of long-lasting protection against the disease or the infection. This review draws insights from previous knowledge regarding the nature and longevity of immunity to the related virus, SARS-CoV, to fill the gaps in our understanding of the immune response to SARS-CoV-2. Deciphering the immunological characteristics that give rise to protective immunity against SARS-CoV-2 is critical to guiding vaccine development and also predicting the course of the pandemic. Here we discuss the recent evidence that characterises the adaptive immune response against SARS-CoV-2 and its potential implications for the generation of memory responses and long-term protection.
Collapse
Affiliation(s)
- David S. Kim
- Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Sarah Rowland-Jones
- Viral Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ester Gea-Mallorquí
- Viral Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
49
|
Taefehshokr N, Taefehshokr S, Heit B. Mechanisms of Dysregulated Humoral and Cellular Immunity by SARS-CoV-2. Pathogens 2020; 9:E1027. [PMID: 33302366 PMCID: PMC7762606 DOI: 10.3390/pathogens9121027] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 11/30/2020] [Accepted: 12/06/2020] [Indexed: 02/06/2023] Open
Abstract
The current coronavirus disease 2019 (COVID-19) pandemic, a disease caused by severe acute respiratory syndrome corona virus 2 (SARS-CoV-2), was first identified in December 2019 in China, and has led to thousands of mortalities globally each day. While the innate immune response serves as the first line of defense, viral clearance requires activation of adaptive immunity, which employs B and T cells to provide sanitizing immunity. SARS-CoV-2 has a potent arsenal of mechanisms used to counter this adaptive immune response through processes, such as T cells depletion and T cell exhaustion. These phenomena are most often observed in severe SARS-CoV-2 patients, pointing towards a link between T cell function and disease severity. Moreover, neutralizing antibody titers and memory B cell responses may be short lived in many SARS-CoV-2 patients, potentially exposing these patients to re-infection. In this review, we discuss our current understanding of B and T cells immune responses and activity in SARS-CoV-2 pathogenesis.
Collapse
Affiliation(s)
- Nima Taefehshokr
- Department of Microbiology and Immunology, Center for Human Immunology, The University of Western Ontario, London, ON N0M 2N0, Canada;
| | - Sina Taefehshokr
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51368, Iran;
| | - Bryan Heit
- Department of Microbiology and Immunology, Center for Human Immunology, The University of Western Ontario, London, ON N0M 2N0, Canada;
- Robarts Research Institute, London, ON N6A 5K8, Canada
| |
Collapse
|
50
|
Cloutier M, Nandi M, Ihsan AU, Chamard HA, Ilangumaran S, Ramanathan S. ADE and hyperinflammation in SARS-CoV2 infection- comparison with dengue hemorrhagic fever and feline infectious peritonitis. Cytokine 2020; 136:155256. [PMID: 32866898 PMCID: PMC7439999 DOI: 10.1016/j.cyto.2020.155256] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/05/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022]
Abstract
The COVID-19 pandemic has rapidly spread around the world with significant morbidity and mortality in a subset of patients including the elderly. The poorer outcomes are associated with 'cytokine storm-like' immune responses, otherwise referred to as 'hyperinflammation'. While most of the infected individuals show minimal or no symptoms and recover spontaneously, a small proportion of the patients exhibit severe symptoms characterized by extreme dyspnea and low tissue oxygen levels, with extensive damage to the lungs referred to as acute respiratory distress symptom (ARDS). The consensus is that the hyperinflammatory response of the host is akin to the cytokine storm observed during sepsis and is the major cause of death. Uncertainties remain on the factors that lead to hyperinflammatory response in some but not all individuals. Hyperinflammation is a common feature in different viral infections such as dengue where existing low-titer antibodies to the virus enhances the infection in immune cells through a process called antibody-dependent enhancement or ADE. ADE has been reported following vaccination or secondary infections with other corona, Ebola and dengue virus. Detailed analysis has shown that antibodies to any viral epitope can induce ADE when present in sub-optimal titers or is of low affinity. In this review we will discuss ADE in the context of dengue and coronavirus infections including Covid-19.
Collapse
Affiliation(s)
- Maryse Cloutier
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Madhuparna Nandi
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Awais Ullah Ihsan
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Hugues Allard Chamard
- Division of Rheumatology, Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Centre de Recherche Clinique, Centre Hospitalier d'Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Subburaj Ilangumaran
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Centre de Recherche Clinique, Centre Hospitalier d'Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Sheela Ramanathan
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Centre de Recherche Clinique, Centre Hospitalier d'Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|