51
|
Islas-Vazquez L, Alvarado-Alvarado YC, Cruz-Aguilar M, Velazquez-Soto H, Villalobos-Gonzalez E, Ornelas-Hall G, Perez-Tapia SM, Jimenez-Martinez MC. Evaluation of the Abdala Vaccine: Antibody and Cellular Response to the RBD Domain of SARS-CoV-2. Vaccines (Basel) 2023; 11:1787. [PMID: 38140191 PMCID: PMC10748004 DOI: 10.3390/vaccines11121787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
Abdala is a recently released RBD protein subunit vaccine against SARS-CoV-2. A few countries, including Mexico, have adopted Abdala as a booster dose in their COVID-19 vaccination schemes. Despite that, most of the Mexican population has received full-scheme vaccination with platforms other than Abdala; little is known regarding Abdala's immunological features, such as its antibody production and T- and B-cell-specific response induction. This work aimed to study antibody production and the adaptive cellular response in the Mexican population that received the Abdala vaccine as a booster. We recruited 25 volunteers and evaluated their RBD-specific antibody production, T- and B-cell-activating profiles, and cytokine production. Our results showed that the Abdala vaccine increases the concentration of RBD IgG-specific antibodies. Regarding the cellular response, after challenging peripheral blood cultures with RBD, the plasmablast (CD19+CD27+CD38High) and transitional B-cell (CD19+CD21+CD38High) percentages increased significantly, while T cells showed an increased activated phenotype (CD3+CD4+CD25+CD69+ and CD3+CD4+CD25+HLA-DR+). Also, IL-2 and IFN-γ increased significantly in the supernatant of the RBD-stimulated cells. Our results suggest that Abdala vaccination, used as a booster, evokes antibody production and the activation of previously generated memory against the SARS-CoV-2 RBD domain.
Collapse
Affiliation(s)
- Lorenzo Islas-Vazquez
- Department of Immunology and Research Unit, Institute of Ophthalmology “Conde de Valenciana Foundation”, Mexico City 06800, Mexico; (L.I.-V.)
| | - Yan Carlos Alvarado-Alvarado
- Department of Immunology and Research Unit, Institute of Ophthalmology “Conde de Valenciana Foundation”, Mexico City 06800, Mexico; (L.I.-V.)
| | - Marisa Cruz-Aguilar
- Department of Immunology and Research Unit, Institute of Ophthalmology “Conde de Valenciana Foundation”, Mexico City 06800, Mexico; (L.I.-V.)
| | - Henry Velazquez-Soto
- Department of Immunology and Research Unit, Institute of Ophthalmology “Conde de Valenciana Foundation”, Mexico City 06800, Mexico; (L.I.-V.)
| | - Eduardo Villalobos-Gonzalez
- Unidad de Vigilancia Epidemiológica Hospitalaria, Institute of Ophthalmology “Conde de Valenciana Foundation”, Mexico City 06800, Mexico
| | - Gloria Ornelas-Hall
- Unidad de Vigilancia Epidemiológica Hospitalaria, Institute of Ophthalmology “Conde de Valenciana Foundation”, Mexico City 06800, Mexico
| | - Sonia Mayra Perez-Tapia
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
- Laboratorio Nacional para Servicios Especializados de Investigación, Desarrollo e Innovación (I+D+i) para Farmoquímicos y Biotecnológicos, LANSEIDI-FarBiotec-CONACyT, Mexico City 11340, Mexico
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (ENCB-IPN), Mexico City 11340, Mexico
| | - Maria C. Jimenez-Martinez
- Department of Immunology and Research Unit, Institute of Ophthalmology “Conde de Valenciana Foundation”, Mexico City 06800, Mexico; (L.I.-V.)
- Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico
| |
Collapse
|
52
|
Recanatini C, GeurtsvanKessel CH, Pas SD, Broens EM, Maas M, van Mansfeld R, Mutsaers-van Oudheusden AJG, van Rijen M, Schippers EF, Stegeman A, Tami A, Veldkamp KE, Visser H, Voss A, Wegdam-Blans MCA, Wertheim HFL, Wever PC, Koopmans MPG, Kluytmans JAJW, Kluytmans-van den Bergh MFQ. Seroprevalence of SARS-CoV-2 antibodies among healthcare workers in Dutch hospitals after the 2020 first wave: a multicentre cross-sectional study with prospective follow-up. Antimicrob Resist Infect Control 2023; 12:137. [PMID: 38031155 PMCID: PMC10688070 DOI: 10.1186/s13756-023-01324-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 10/22/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND We aimed to estimate the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) seroprevalence and describe its determinants and associated symptoms among unvaccinated healthcare workers (HCWs) after the first wave of the pandemic. METHODS HCWs from 13 Dutch hospitals were screened for antibodies against the spike protein of SARS-CoV-2 in June-July 2020 and after three months. Participants completed a retrospective questionnaire on determinants for occupational and community exposure to SARS-CoV-2 and symptoms suggestive of COVID-19 experienced since January 2020. The seroprevalence was calculated per baseline characteristic and symptom at baseline and after follow-up. Adjusted odds ratios (aOR) for seropositivity were determined using logistic regression. RESULTS Among 2328 HCWs, 323 (13.9%) were seropositive at enrolment, 49 of whom (15%) reported no previous symptoms suggestive of COVID-19. During follow-up, only 1% of the tested participants seroconverted. Seroprevalence was higher in younger HCWs compared to the mid-age category (aOR 1.53, 95% CI 1.07-2.18). Nurses (aOR 2.21, 95% CI 1.34-3.64) and administrative staff (aOR 1.87, 95% CI 1.02-3.43) had a higher seroprevalence than physicians. The highest seroprevalence was observed in HCWs in the emergency department (ED) (aOR 1.79, 95% CI 1.10-2.91), the lowest in HCWs in the intensive, high, or medium care units (aOR 0.47, 95% CI 0.31-0.71). Chronic respiratory disease, smoking, and having a dog were independently associated with a lower seroprevalence, while HCWs with diabetes mellitus had a higher seroprevalence. In a multivariable model containing all self-reported symptoms since January 2020, altered smell and taste, fever, general malaise/fatigue, and muscle aches were positively associated with developing antibodies, while sore throat and chills were negatively associated. CONCLUSIONS The SARS-CoV-2 seroprevalence in unvaccinated HCWs of 13 Dutch hospitals was 14% in June-July 2020 and remained stable after three months. A higher seroprevalence was observed in the ED and among nurses, administrative and young staff, and those with diabetes mellitus, while a lower seroprevalence was found in HCWs in intensive, high, or medium care, and those with self-reported lung disease, smokers, and dog owners. A history of altered smell or taste, fever, muscle aches and fatigue were independently associated with the presence of SARS-CoV-2 antibodies in unvaccinated HCWs.
Collapse
Affiliation(s)
- Claudia Recanatini
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| | | | - Suzan D Pas
- Microvida Laboratory for Medical Microbiology, Bravis Hospital, Roosendaal, The Netherlands
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Els M Broens
- Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Martje Maas
- Department of Internal Medicine, Bernhoven Hospital, Uden, The Netherlands
| | - Rosa van Mansfeld
- Department of Medical Microbiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Miranda van Rijen
- Department of Infection Control, Amphia Hospital, Breda, The Netherlands
| | - Emile F Schippers
- Department of Internal Medicine, Haga Hospital, The Hague, The Netherlands
| | - Arjan Stegeman
- Department of Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Adriana Tami
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Karin Ellen Veldkamp
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hannah Visser
- Department of Internal Medicine, Beatrix Hospital, Gorinchem, The Netherlands
| | - Andreas Voss
- Department of Medical Microbiology and Infectious Diseases, Canisius Wilhelmina Hospital, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marjolijn C A Wegdam-Blans
- Catharina Hospital, Eindhoven, The Netherlands
- Hospital St. Jans Gasthuis, Weert, The Netherlands
- Department of Medical Microbiology, Stichting PAMM, Veldhoven, The Netherlands
| | - Heiman F L Wertheim
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter C Wever
- Department of Medical Microbiology and Infection Control, Jeroen Bosch Hospital, 's Hertogenbosch, The Netherlands
| | - Marion P G Koopmans
- Viroscience Department, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jan A J W Kluytmans
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Infection Control, Amphia Hospital, Breda, The Netherlands
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marjolein F Q Kluytmans-van den Bergh
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Infection Control, Amphia Hospital, Breda, The Netherlands
- Amphia Academy Infectious Disease Foundation, Amphia Hospital, Breda, The Netherlands
| |
Collapse
|
53
|
Bešević J, Lacey B, Callen H, Omiyale W, Conroy M, Feng Q, Crook DW, Doherty N, Ebner D, Eyre DW, Fry D, Horn E, Jones EY, Marsden BD, Peto TEA, Starkey F, Stuart D, Welsh S, Wood N, Young A, Young A, Effingham M, Collins R, Holliday J, Allen N. Persistence of SARS-CoV-2 antibodies over 18 months following infection: UK Biobank COVID-19 Serology Study. J Epidemiol Community Health 2023; 78:jech-2023-220569. [PMID: 37923370 PMCID: PMC10850672 DOI: 10.1136/jech-2023-220569] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 10/08/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND Little is known about the persistence of antibodies after the first year following SARS-CoV-2 infection. We aimed to determine the proportion of individuals that maintain detectable levels of SARS-CoV-2 antibodies over an 18-month period following infection. METHODS Population-based prospective study of 20 000 UK Biobank participants and their adult relatives recruited in May 2020. The proportion of SARS-CoV-2 cases testing positive for immunoglobulin G (IgG) antibodies against the spike protein (IgG-S), and the nucleocapsid protein (IgG-N), was calculated at varying intervals following infection. RESULTS Overall, 20 195 participants were recruited. Their median age was 56 years (IQR 39-68), 56% were female and 88% were of white ethnicity. The proportion of SARS-CoV-2 cases with IgG-S antibodies following infection remained high (92%, 95% CI 90%-93%) at 6 months after infection. Levels of IgG-N antibodies following infection gradually decreased from 92% (95% CI 88%-95%) at 3 months to 72% (95% CI 70%-75%) at 18 months. There was no strong evidence of heterogeneity in antibody persistence by age, sex, ethnicity or socioeconomic deprivation. CONCLUSION This study adds to the limited evidence on the long-term persistence of antibodies following SARS-CoV-2 infection, with likely implications for waning immunity following infection and the use of IgG-N in population surveys.
Collapse
Affiliation(s)
- Jelena Bešević
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| | - Ben Lacey
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| | - Howard Callen
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| | - Wemimo Omiyale
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| | - Megan Conroy
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| | - Qi Feng
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| | - Derrick W Crook
- Nuffield Department of Medicine (NDM), University of Oxford, Oxford, UK
| | | | - Daniel Ebner
- Nuffield Department of Medicine (NDM), University of Oxford, Oxford, UK
| | - David W Eyre
- University of Oxford Big Data Institute, Oxford, UK
| | | | - Edward Horn
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| | - E Yvonne Jones
- Nuffield Department of Medicine (NDM), University of Oxford, Oxford, UK
| | - Brian D Marsden
- Nuffield Department of Medicine (NDM), University of Oxford, Oxford, UK
| | - Tim E A Peto
- Nuffield Department of Medicine (NDM), University of Oxford, Oxford, UK
| | - Fenella Starkey
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| | - David Stuart
- Nuffield Department of Medicine (NDM), University of Oxford, Oxford, UK
| | | | - Natasha Wood
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| | - Alan Young
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
- UK Biobank, Stockport, UK
| | - Allen Young
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| | | | - Rory Collins
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
- UK Biobank, Stockport, UK
| | - Jo Holliday
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| | - Naomi Allen
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
- UK Biobank, Stockport, UK
| |
Collapse
|
54
|
Kenny G, O'Reilly S, Wrigley Kelly N, Negi R, Gaillard C, Alalwan D, Saini G, Alrawahneh T, Francois N, Angeliadis M, Garcia Leon AA, Tinago W, Feeney ER, Cotter AG, de Barra E, Yousif O, Horgan M, Doran P, Stemler J, Koehler P, Cox RJ, O'Shea D, Olesen OF, Landay A, Hogan AE, Lelievre JD, Gautier V, Cornely OA, Mallon PWG. Distinct receptor binding domain IgG thresholds predict protective host immunity across SARS-CoV-2 variants and time. Nat Commun 2023; 14:7015. [PMID: 37919289 PMCID: PMC10622572 DOI: 10.1038/s41467-023-42717-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/19/2023] [Indexed: 11/04/2023] Open
Abstract
SARS-CoV-2 neutralising antibodies provide protection against COVID-19. Evidence from early vaccine trials suggested binding antibody thresholds could serve as surrogate markers of neutralising capacity, but whether these thresholds predict sufficient neutralising capacity against variants of concern (VOCs), and whether this is impacted by vaccine or infection history remains unclear. Here we analyse individuals recovered from, vaccinated or with hybrid immunity against SARS-CoV-2. An NT50 ≥ 100 IU confers protection in vaccine trials, however, as VOC induce a reduction in NT50, we use NT50 ≥ 1000 IU as a cut off for WT NT50 that would retain neutralisation against VOC. In unvaccinated convalescent participants, a receptor binding domain (RBD) IgG of 456 BAU/mL predicts an NT50 against WT of 1000 IU with an accuracy of 80% (95%CI 73-86%). This threshold maintains accuracy in determining loss of protective immunity against VOC in two vaccinated cohorts. It predicts an NT50 < 100 IU against Beta with an accuracy of 80% (95%CI 67-89%) in 2 vaccine dose recipients. In booster vaccine recipients with a history of COVID-19 (hybrid immunity), accuracy is 87% (95%CI 77-94%) in determining an NT50 of <100 IU against BA.5. This analysis provides a discrete threshold that could be used in future clinical studies.
Collapse
Affiliation(s)
- Grace Kenny
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland.
| | - Sophie O'Reilly
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
| | - Neil Wrigley Kelly
- St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co Kildare, Ireland
| | - Riya Negi
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
| | - Colette Gaillard
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
| | - Dana Alalwan
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
| | - Gurvin Saini
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
| | - Tamara Alrawahneh
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
| | - Nathan Francois
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
| | - Matthew Angeliadis
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
| | - Alejandro Abner Garcia Leon
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
| | - Willard Tinago
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
| | - Eoin R Feeney
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - Aoife G Cotter
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- Department of Infectious Diseases, Mater Misericordiae University Hospital, Eccles St, Dublin 7, Ireland
| | - Eoghan de Barra
- Department of Infectious Diseases, Beaumont Hospital, Beaumont, Dublin 9, Ireland
- Department of International Health and Tropical Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Obada Yousif
- Endocrinology Department, Wexford General Hospital, Carricklawn, Wexford, Ireland
| | - Mary Horgan
- Department of Infectious Diseases, Cork University Hospital, Wilton, Co Cork, Ireland
| | - Peter Doran
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Jannik Stemler
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Internal Medicine and University of Cologne, Faculty of Medicine Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne Department Cologne, Cologne, Germany
| | - Philipp Koehler
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Internal Medicine and University of Cologne, Faculty of Medicine Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne Department Cologne, Cologne, Germany
| | - Rebecca Jane Cox
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Donal O'Shea
- St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - Ole F Olesen
- European Vaccine Initiative, Heidelberg, Germany
| | - Alan Landay
- Department of internal Medicine, Rush University, Chicago, IL, USA
| | - Andrew E Hogan
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co Kildare, Ireland
- National Children's Research Centre, Dublin 12, Ireland
| | | | - Virginie Gautier
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
| | - Oliver A Cornely
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Internal Medicine and University of Cologne, Faculty of Medicine Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne Department Cologne, Cologne, Germany
| | - Patrick W G Mallon
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| |
Collapse
|
55
|
Guan X, Huang Q, Dong M, Li M, Xie H, Wei X, Kang L, Wang X, Li A, Wang Q, Huang F, Wang Q. SARS-CoV-2-specific antibody and T-cell immunity in convalescents after infection wave in Beijing in late 2022. J Infect 2023; 87:413-419. [PMID: 37652314 DOI: 10.1016/j.jinf.2023.08.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/24/2023] [Accepted: 08/27/2023] [Indexed: 09/02/2023]
Abstract
OBJECTIVES To evaluate SARS-CoV-2-specific antibody and T-cell responses in convalescents 5 months after infection wave in Beijing from December 2022 to January 2023 to prevent reinfection and severe disease. METHODS Convalescents and uninfected individuals vaccinated with different doses were enrolled to assess the IFNγ T-cell responses against SARS-CoV-2 prototype strain, BF.7, BQ.1, and XBB. Neutralizing antibodies against prototype strain, BF.7, BA.5, and XBB and immunoglobulin G antibody were further analyzed. RESULTS In convalescents, the IFNγ T-cell response was significantly higher than that of uninfected individuals (all P < 0.001), and the T-cell response against XBB had no significant difference from that of SARS-CoV-2 prototype strain and BF.7 and BQ.1 (all P > 0.05). The seropositive rates of IgG antibodies were 100% (303/303) with a median concentration of 90.52 (95% CI, 82.52-99.37). The neutralizing antibodies titers of convalescents against BF.7 and BA.5 were higher than that against the prototype strain (both P < 0.001), while XBB.1.5 was lower (P < 0.001). T-cell response, IgG and neutralizing antibodies had no significant difference in convalescents vaccinated with different doses (all P > 0.05). CONCLUSIONS The immunities may have some protective effect against possible future outbreaks and severe diseases of COVID-19.
Collapse
Affiliation(s)
- Xuejiao Guan
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China
| | - Qi Huang
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China; School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China
| | - Mei Dong
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China
| | - Maozhong Li
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China
| | - Hui Xie
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China
| | - Xiaofeng Wei
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China; School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China
| | - Lu Kang
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China
| | - Xue Wang
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China
| | - Aihua Li
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China
| | - Qing Wang
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China; School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China
| | - Fang Huang
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China; Beijing Research Center for Respiratory Infectious Diseases, People's Republic of China.
| | - Quanyi Wang
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, People's Republic of China; Beijing Research Center for Respiratory Infectious Diseases, People's Republic of China.
| |
Collapse
|
56
|
Corsini CA, Filgueiras PS, Almeida NB, Miranda DAD, Gomes SV, Lourenço AJ, Bicalho CM, Assis JVD, Amorim RN, Silva RA, Vilela RV, Lima TM, Abreu DPD, Alvim RG, Castilho LR, Martins-Filho OA, Otta DA, Grenfell RF. Antibody response and soluble mediator profile in the first six months following acute SARS-CoV-2 infection. Sci Rep 2023; 13:18606. [PMID: 37903875 PMCID: PMC10616118 DOI: 10.1038/s41598-023-43263-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/21/2023] [Indexed: 11/01/2023] Open
Abstract
The COVID-19 pandemic has caused a severe global health and economic crisis, with significant consequences for human mortality and morbidity. Therefore, there is an urgent need for more studies on the immune response to SARS-CoV-2 infection, both to enhance its effectiveness and prevent its deleterious effects. This study presents the chronology of antibodies during six months after infection in hospitalized patients and the kinetics of serum soluble mediators of the cellular response triggered by SARS-CoV-2. Samples and clinical data from 330 patients hospitalized at the Hospital da Baleia in Belo Horizonte, Brazil, who were suspected of having COVID-19, were collected at the time of hospitalization and during 6 months after infection. The immune response was analyzed by enzyme-linked immunosorbent assay (ELISA) and flow cytometry. There was a significant difference in IgM specific antibody titers from the 7th to 60th days after infection between COVID-19 negative and positive patients. Soon after 60 days after infection, antibody levels started to reduce, becoming similar to the antibody levels of the COVID-19 negative patients. IgG specific antibodies started to be detectable after 9 days of infection and antibody levels were comparatively higher in positive patients as soon as after 7 days. Furthermore, IgG levels remained higher in these patients during the complete period of 180 days after infection. The study observed similar antibody profiles between different patient groups. The soluble systemic biomarkers evaluated showed a decrease during the six months after hospitalization, except for CCL11, CXCL8, CCL3, CCL4, CCL5, IL-6, IFN-g, IL-17, IL-5, FGF-basic, PDGF, VEGF, G-CSF, and GM-CSF. The results indicate that IgM antibodies are more prominent in the early stages of infection, while IgG antibodies persist for a longer period. Additionally, the study identified that patients with COVID-19 have elevated levels of biomarkers after symptom onset, which decrease over time.
Collapse
Affiliation(s)
- Camila A Corsini
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (FIOCRUZ), 1715 Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Priscilla S Filgueiras
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (FIOCRUZ), 1715 Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
- Department of Pathology, College of Medicine, Federal University of Minas Gerais, 6627 Avenida Presidente Antônio Carlos, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Nathalie Bf Almeida
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (FIOCRUZ), 1715 Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Daniel Ap de Miranda
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (FIOCRUZ), 1715 Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Sarah Vc Gomes
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (FIOCRUZ), 1715 Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Adelina Junia Lourenço
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (FIOCRUZ), 1715 Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
- Hospital da Baleia, Benjamin Guimarães Foundation, 1464 Juramento Street, Belo Horizonte, Minas Gerais, 30285-408, Brazil
| | - Cecilia Mf Bicalho
- Hospital da Baleia, Benjamin Guimarães Foundation, 1464 Juramento Street, Belo Horizonte, Minas Gerais, 30285-408, Brazil
| | - Jessica V de Assis
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (FIOCRUZ), 1715 Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
- Department of Pathology, College of Medicine, Federal University of Minas Gerais, 6627 Avenida Presidente Antônio Carlos, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Raquel Nh Amorim
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (FIOCRUZ), 1715 Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Raphael A Silva
- Hospital da Baleia, Benjamin Guimarães Foundation, 1464 Juramento Street, Belo Horizonte, Minas Gerais, 30285-408, Brazil
| | - Raquel Vr Vilela
- Hospital da Baleia, Benjamin Guimarães Foundation, 1464 Juramento Street, Belo Horizonte, Minas Gerais, 30285-408, Brazil
| | - Tulio M Lima
- Cell Culture Engineering Laboratory, COPPE, Universidade Federal do Rio de Janeiro, 550 Pedro Calmon Avenue, Rio de Janeiro, Rio de Janeiro, 21941-598, Brazil
| | - Daniel Pb de Abreu
- Cell Culture Engineering Laboratory, COPPE, Universidade Federal do Rio de Janeiro, 550 Pedro Calmon Avenue, Rio de Janeiro, Rio de Janeiro, 21941-598, Brazil
| | - Renata Gf Alvim
- Cell Culture Engineering Laboratory, COPPE, Universidade Federal do Rio de Janeiro, 550 Pedro Calmon Avenue, Rio de Janeiro, Rio de Janeiro, 21941-598, Brazil
| | - Leda R Castilho
- Cell Culture Engineering Laboratory, COPPE, Universidade Federal do Rio de Janeiro, 550 Pedro Calmon Avenue, Rio de Janeiro, Rio de Janeiro, 21941-598, Brazil
| | - Olindo A Martins-Filho
- Grupo Integrado de Pesquisa em Biomarcadores, Oswaldo Cruz Foundation (FIOCRUZ), 1715 Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Dayane A Otta
- Grupo Integrado de Pesquisa em Biomarcadores, Oswaldo Cruz Foundation (FIOCRUZ), 1715 Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Rafaella Fq Grenfell
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (FIOCRUZ), 1715 Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil.
- Department of Pathology, College of Medicine, Federal University of Minas Gerais, 6627 Avenida Presidente Antônio Carlos, Belo Horizonte, Minas Gerais, 31270-901, Brazil.
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA, 30602-7387, USA.
| |
Collapse
|
57
|
Doherty J, O'Morain N, Stack R, Tosetto M, Inzitiari R, O'Reilly S, Gu L, Sheridan J, Cullen G, Mc Dermott E, Buckley M, Horgan G, Mulcahy H, Walshe M, Ryan EJ, Gautier V, Prostko J, Frias E, Daghfal D, Doran P, O'Morain C, Doherty GA. Reduced Serological Response to COVID-19 Booster Vaccine is Associated with Reduced B Cell Memory in Patients With Inflammatory Bowel Disease; VARIATION [VAriability in Response in IBD AgainsT SARS-COV-2 ImmunisatiON]. J Crohns Colitis 2023; 17:1445-1456. [PMID: 37018462 DOI: 10.1093/ecco-jcc/jjad065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Indexed: 04/07/2023]
Abstract
BACKGROUND AND AIMS Patients with inflammatory bowel disease [IBD] have an attenuated response to initial COVID-19 vaccination. We sought to characterize the impact of IBD and its treatment on responses after the third vaccine against SARS-CoV-2. METHODS This was a prospective multicentre observational study of patients with IBD [n = 202] and healthy controls [HC, n = 92]. Serological response to vaccination was assessed by quantification of anti-spike protein [SP] immunoglobulin [Ig]G levels [anti-SPIgG] and in vitro neutralization of binding to angiotensin-converting enzyme 2 [ACE2]. Peripheral blood B-cell phenotype populations were assessed by flow cytometry. SARS-CoV-2 antigen-specific B-cell responses were assessed in ex vivo culture. RESULTS Median anti-SP IgG post-third vaccination in our IBD cohort was significantly lower than HCs [7862 vs 19 622 AU/mL, p < 0.001] as was ACE2 binding inhibition [p < 0.001]. IBD patients previously infected with COVID-19 [30%] had similar quantitative antibody response as HCs previously infected with COVID-19 [p = 0.12]. Lowest anti-SP IgG titres and neutralization were seen in IBD patients on anti-tumour necrosis factor [anti-TNF] agents, without prior COVID-19 infection, but all IBD patients show an attenuated vaccine response compared to HCs. Patients with IBD have reduced memory B-cell populations and attenuated B-cell responses to SARS-CoV-2 antigens if not previously infected with COVID-19 [p = 0.01]. Higher anti-TNF drug levels and zinc levels <65 ng/ml were associated with significantly lower serological responses. CONCLUSIONS Patients with IBD have an attenuated response to three doses of SARS-CoV-2 vaccine. Physicians should consider patients with higher anti-TNF drug levels and/or zinc deficiency as potentially at higher risk of attenuated response to vaccination.
Collapse
Affiliation(s)
- Jayne Doherty
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
- INITIative IBD Research Network (www.initiativeibd.ie)
| | - Neil O'Morain
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Roisin Stack
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Miriam Tosetto
- School of Medicine, University College Dublin, Dublin, Ireland
| | | | - Sophie O'Reilly
- Centre for Experimental Pathogen Host Research, School of Medicine, University College Dublin, Ireland
| | - Lili Gu
- Centre for Experimental Pathogen Host Research, School of Medicine, University College Dublin, Ireland
| | - Juliette Sheridan
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Garret Cullen
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
- INITIative IBD Research Network (www.initiativeibd.ie)
| | - Edel Mc Dermott
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
| | - Maire Buckley
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- St Michaels Hospital, Dun Laoghaire, Co Dublin, Ireland
| | - Gareth Horgan
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- St Columcille's Hospital, Loughlinstown, Co Dublin, Ireland
| | - Hugh Mulcahy
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Margaret Walshe
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
- INITIative IBD Research Network (www.initiativeibd.ie)
| | - Elizabeth J Ryan
- Department of Biological Sciences, Health Research Institute, University of Limerick, Limerick, Ireland
| | - Virginie Gautier
- Centre for Experimental Pathogen Host Research, School of Medicine, University College Dublin, Ireland
| | - John Prostko
- Abbott Laboratories, Abbott Diagnostics, Lake Forest, IL 60045, USA
| | - Edwin Frias
- Abbott Laboratories, Abbott Diagnostics, Lake Forest, IL 60045, USA
| | - David Daghfal
- Abbott Laboratories, Abbott Diagnostics, Lake Forest, IL 60045, USA
| | - Peter Doran
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Colm O'Morain
- Beacon Hospital, Sandyford, Co. Dublin and Trinity College Dublin, Ireland
- INITIative IBD Research Network (www.initiativeibd.ie)
| | - Glen A Doherty
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
- INITIative IBD Research Network (www.initiativeibd.ie)
| |
Collapse
|
58
|
Peterhoff D, Wiegrebe S, Einhauser S, Patt AJ, Beileke S, Günther F, Steininger P, Niller HH, Burkhardt R, Küchenhoff H, Gefeller O, Überla K, Heid IM, Wagner R. Population-based study of the durability of humoral immunity after SARS-CoV-2 infection. Front Immunol 2023; 14:1242536. [PMID: 37868969 PMCID: PMC10585261 DOI: 10.3389/fimmu.2023.1242536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 09/19/2023] [Indexed: 10/24/2023] Open
Abstract
SARS-CoV-2 antibody quantity and quality are key markers of humoral immunity. However, there is substantial uncertainty about their durability. We investigated levels and temporal change of SARS-CoV-2 antibody quantity and quality. We analyzed sera (8 binding, 4 avidity assays for spike-(S-)protein and nucleocapsid-(N-)protein; neutralization) from 211 seropositive unvaccinated participants, from the population-based longitudinal TiKoCo study, at three time points within one year after infection with the ancestral SARS-CoV-2 virus. We found a significant decline of neutralization titers and binding antibody levels in most assays (linear mixed regression model, p<0.01). S-specific serum avidity increased markedly over time, in contrast to N-specific. Binding antibody levels were higher in older versus younger participants - a difference that disappeared for the asymptomatic-infected. We found stronger antibody decline in men versus women and lower binding and avidity levels in current versus never-smokers. Our comprehensive longitudinal analyses across 13 antibody assays suggest decreased neutralization-based protection and prolonged affinity maturation within one year after infection.
Collapse
Affiliation(s)
- David Peterhoff
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Simon Wiegrebe
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
- Statistical Consulting Unit StaBLab, Department of Statistics, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Sebastian Einhauser
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
| | - Arisha J. Patt
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Stephanie Beileke
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Felix Günther
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
- Statistical Consulting Unit StaBLab, Department of Statistics, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Philipp Steininger
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Hans H. Niller
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Helmut Küchenhoff
- Statistical Consulting Unit StaBLab, Department of Statistics, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Olaf Gefeller
- Department of Medical Informatics, Biometry and Epidemiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Klaus Überla
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Iris M. Heid
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
59
|
Abdullahi A, Frimpong J, Cheng MTK, Aliyu SH, Smith C, Abimiku A, Phillips RO, Owusu M, Gupta RK. Performance of SARS COV-2 IgG Anti-N as an Independent Marker of Exposure to SARS COV-2 in an Unvaccinated West African Population. Am J Trop Med Hyg 2023; 109:890-894. [PMID: 37580023 PMCID: PMC10551093 DOI: 10.4269/ajtmh.23-0179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/22/2023] [Indexed: 08/16/2023] Open
Abstract
Determination of previous SARS-COV-2 infection is hampered by the absence of a standardized test. The marker used to assess previous exposure is IgG antibody to the nucleocapsid (IgG anti-N), although it is known to wane quickly from peripheral blood. The accuracies of seven antibody tests (virus neutralization test, IgG anti-N, IgG anti-spike [anti-S], IgG anti-receptor binding domain [anti-RBD], IgG anti-N + anti-RBD, IgG anti-N + anti-S, and IgG anti-S + anti-RBD), either singly or in combination, were evaluated on 502 cryopreserved serum samples collected before the COVID-19 vaccination rollout in Kumasi, Ghana. The accuracy of each index test was measured using a composite reference standard based on a combination of neutralization test and IgG anti-N antibody tests. According to the composite reference, 262 participants were previously exposed; the most sensitive test was the virus neutralization test, with 95.4% sensitivity (95% CI: 93.6-97.3), followed by 79.0% for IgG anti-N + anti-S (95% CI: 76.3-83.3). The most specific tests were virus neutralization and IgG anti-N, both with 100% specificity. Viral neutralization and IgG anti-N + anti-S were the overall most accurate tests, with specificity/sensitivity of 100/95.2% and 79.0/92.1%, respectively. Our findings indicate that IgG anti-N alone is an inadequate marker of prior exposure to SARS COV-2 in this population. Virus neutralization assay appears to be the most accurate assay in discerning prior infection. A combination of IgG anti-N and IgG anti-S is also accurate and suited for assessment of SARS COV-2 exposure in low-resource settings.
Collapse
Affiliation(s)
- Adam Abdullahi
- Cambridge Institute of Therapeutic Immunology & Infectious Disease, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Institute of Human Virology, Abuja, Nigeria
| | - James Frimpong
- Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Kumasi Centre for Collaborative Research in Tropical Medicine, Kumasi, Ghana
| | - Mark T. K. Cheng
- Cambridge Institute of Therapeutic Immunology & Infectious Disease, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Sani H. Aliyu
- Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | | | | | - Richard Odame Phillips
- Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Kumasi Centre for Collaborative Research in Tropical Medicine, Kumasi, Ghana
| | - Michael Owusu
- Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Kumasi Centre for Collaborative Research in Tropical Medicine, Kumasi, Ghana
| | - Ravindra K. Gupta
- Cambridge Institute of Therapeutic Immunology & Infectious Disease, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Africa Health Research Institute, Durban, South Africa
| |
Collapse
|
60
|
Shin SS, Bender M, Malherbe DC, Vasquez H, Doratt BM, Messaoudi I. SARS-Cov-2 Infection and Seroconversion Rates in Healthcare Providers Prior to COVID-19 Vaccine Rollout. Biol Res Nurs 2023; 25:505-515. [PMID: 36869766 PMCID: PMC9988593 DOI: 10.1177/10998004231161632] [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] [Indexed: 03/05/2023]
Abstract
OBJECTIVE A 6-month longitudinal surveillance study of asymptomatic healthcare providers (HCP) was carried out at a large urban academic medical center in the United States to assess whether their job occupation with higher exposure risks to SARS-CoV-2 would equate with higher risk of contracting COVID-19 at the beginning of the pandemic before COVID-19 vaccines were available. METHODS A longitudinal cohort study design was used to collect and analyze immunological and virological monitoring data and self-report survey assessments of personal protective equipment (PPE) availability, adherence to infection control guidelines, and time spent on COVID-19 wards. RESULTS Among 289 eligible participants, SARS-CoV-2 exposure risk was high with 48-69% participants working in COVID-19 units and more than 30% of them caring for COVID-19 patients. However, the seroconversion rate was low with only 2.1% of participants developing humoral or cellular immunity against SARS-CoV-2. CONCLUSION Our study findings suggest that, for this HCP cohort working at a large urban academic medical center, a low incidence of SARS-CoV-2 infection could be maintained under conditions of strict infection prevention protocols and reliable PPE availability.
Collapse
Affiliation(s)
- Sanghyuk S. Shin
- Sue & Bill Gross School of Nursing, University of California, Irvine, CA, USA
| | - Miriam Bender
- Sue & Bill Gross School of Nursing, University of California, Irvine, CA, USA
| | - Delphine C. Malherbe
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Hannah Vasquez
- Sue & Bill Gross School of Nursing, University of California, Irvine, CA, USA
| | - Brianna M. Doratt
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Ilhem Messaoudi
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
61
|
Kubisch U, Sandoni A, Wurm J, Schienkiewitz A, Schlaud M, Kuttig T, Finkel B, Jordan S, Loss J. SARS-CoV-2 seroconversion in children attending daycare versus adults in Germany between October 2020 and June 2021. COMMUNICATIONS MEDICINE 2023; 3:124. [PMID: 37714948 PMCID: PMC10504330 DOI: 10.1038/s43856-023-00352-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 08/31/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND Data on seroconversion rates after SARS-CoV-2 infection in young children (<6 years) is scarce. The present study compares seroconversion rates between young children and adults and identifies associated factors. METHODS The COALA study ("Corona-outbreak-related examinations in daycare centers") investigated transmission dynamics of SARS-CoV-2 in daycare centers and associated households (10/2020-06/2021). 114 individuals tested positive for SARS-CoV-2 through PCR either prior to the study period by health authorities or in PCR testing during the study period. Two capillary blood samples were obtained within five weeks consecutively and tested for SARS-CoV-2 IgG-antibodies (second sampling depending on positive PCR). Results from 91 participants (38 young children 1-6 years, 53 adults) were included in the analyses. RESULTS Seroconversion rate in young children is significantly higher than in adults (97.4% versus 66%). High viral load and longer time interval between the probable date of infection and antibody testing are associated with seroconversion. CONCLUSIONS Our findings depict substantial development of specific antibodies in young children after SARS-CoV-2 infection. This may provide temporary protection from re-infection for young children or severe disease for this age group.
Collapse
Affiliation(s)
- Ulrike Kubisch
- Department of Epidemiology and Health Monitoring, Robert Koch Institute, Berlin, Germany.
| | - Anna Sandoni
- Department of Epidemiology and Health Monitoring, Robert Koch Institute, Berlin, Germany
| | - Juliane Wurm
- Department of Epidemiology and Health Monitoring, Robert Koch Institute, Berlin, Germany
| | - Anja Schienkiewitz
- Department of Epidemiology and Health Monitoring, Robert Koch Institute, Berlin, Germany
| | - Martin Schlaud
- Department of Epidemiology and Health Monitoring, Robert Koch Institute, Berlin, Germany
| | - Tim Kuttig
- Department of Epidemiology and Health Monitoring, Robert Koch Institute, Berlin, Germany
| | - Bianca Finkel
- Department of Epidemiology and Health Monitoring, Robert Koch Institute, Berlin, Germany
| | - Susanne Jordan
- Department of Epidemiology and Health Monitoring, Robert Koch Institute, Berlin, Germany
| | - Julika Loss
- Department of Epidemiology and Health Monitoring, Robert Koch Institute, Berlin, Germany
| |
Collapse
|
62
|
Curtis NC, Shin S, Hederman AP, Connor RI, Wieland-Alter WF, Ionov S, Boylston J, Rose J, Sakharkar M, Dorman DB, Dessaint JA, Gwilt LL, Crowley AR, Feldman J, Hauser BM, Schmidt AG, Ashare A, Walker LM, Wright PF, Ackerman ME, Lee J. Characterization of SARS-CoV-2 Convalescent Patients' Serological Repertoire Reveals High Prevalence of Iso-RBD Antibodies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.08.556349. [PMID: 37745524 PMCID: PMC10515772 DOI: 10.1101/2023.09.08.556349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
While our understanding of SARS-CoV-2 pathogenesis and antibody responses following infection and vaccination has improved tremendously since the outbreak in 2019, the sequence identities and relative abundances of the individual constituent antibody molecules in circulation remain understudied. Using Ig-Seq, we proteomically profiled the serological repertoire specific to the whole ectodomain of SARS-CoV-2 prefusion-stabilized spike (S) as well as to the receptor binding domain (RBD) over a 6-month period in four subjects following SARS-CoV-2 infection before SARS-CoV-2 vaccines were available. In each individual, we identified between 59 and 167 unique IgG clonotypes in serum. To our surprise, we discovered that ∼50% of serum IgG specific for RBD did not recognize prefusion-stabilized S (referred to as iso-RBD antibodies), suggesting that a significant fraction of serum IgG targets epitopes on RBD inaccessible on the prefusion-stabilized conformation of S. On the other hand, the abundance of iso-RBD antibodies in nine individuals who received mRNA-based COVID-19 vaccines encoding prefusion-stabilized S was significantly lower (∼8%). We expressed a panel of 12 monoclonal antibodies (mAbs) that were abundantly present in serum from two SARS-CoV-2 infected individuals, and their binding specificities to prefusion-stabilized S and RBD were all in agreement with the binding specificities assigned based on the proteomics data, including 1 iso-RBD mAb which bound to RBD but not to prefusion-stabilized S. 2 of 12 mAbs demonstrated neutralizing activity, while other mAbs were non-neutralizing. 11 of 12 mAbs also bound to S (B.1.351), but only 1 maintained binding to S (B.1.1.529). This particular mAb binding to S (B.1.1.529) 1) represented an antibody lineage that comprised 43% of the individual's total S-reactive serum IgG binding titer 6 months post-infection, 2) bound to the S from a related human coronavirus, HKU1, and 3) had a high somatic hypermutation level (10.9%), suggesting that this antibody lineage likely had been elicited previously by pre-pandemic coronavirus and was re-activated following the SARS-CoV-2 infection. All 12 mAbs demonstrated their ability to engage in Fc-mediated effector function activities. Collectively, our study provides a quantitative overview of the serological repertoire following SARS-CoV-2 infection and the significant contribution of iso-RBD antibodies, demonstrating how vaccination strategies involving prefusion-stabilized S may have reduced the elicitation of iso-RBD serum antibodies which are unlikely to contribute to protection.
Collapse
|
63
|
Pérez-Juárez H, Serrano-Vázquez A, Godínez-Alvarez H, González E, Rojas-Velázquez L, Moran P, Portillo-Bobadilla T, Ramiro M, Hernández E, Lau C, Martínez M, Padilla MDLÁ, Zaragoza ME, Taboada B, Palomares LA, López S, Alagón A, Arias CF, Ximénez C. Longitudinal anti-SARS-CoV-2 antibody immune response in acute and convalescent patients. Front Cell Infect Microbiol 2023; 13:1239700. [PMID: 37743860 PMCID: PMC10515199 DOI: 10.3389/fcimb.2023.1239700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/14/2023] [Indexed: 09/26/2023] Open
Abstract
Despite global efforts to assess the early response and persistence of SARS-CoV-2 antibodies in patients infected with or recovered from COVID-19, our understanding of the factors affecting its dynamics remains limited. This work aimed to evaluate the early and convalescent immunity of outpatients infected with SARS-CoV-2 and to determine the factors that affect the dynamics and persistence of the IgM and IgG antibody response. Seropositivity of volunteers from Mexico City and the State of Mexico, Mexico, was evaluated by ELISA using the recombinant receptor-binding domain (RBD) of the SARS-CoV-2 Spike protein for 90 days, at different time points (1, 15, 45, 60, and 90 days) after molecular diagnosis (RT-qPCR). Gender, age range, body mass index (BMI), comorbidities, and clinical spectrum of disease were analyzed to determine associations with the dynamics of anti-SARS-CoV-2 antibodies. On 90 days post-infection, individuals with moderate and asymptomatic disease presented the lowest levels of IgM, while for IgG, at the same time, the highest levels occurred with mild and moderate disease. The IgM and IgG levels were related to the clinical spectrum of disease, BMI, and the presence/absence of comorbidities through regression trees. The results suggest that the dynamics of anti-SARS-CoV-2 IgM and IgG antibodies in outpatients could be influenced by the clinical spectrum of the disease. In addition, the persistence of antibodies against SARS-CoV-2 could be related to the clinical spectrum of the disease, BMI, and the presence/absence of comorbidities.
Collapse
Affiliation(s)
- Horacio Pérez-Juárez
- Laboratorio de Inmunología, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Estancias Posdoctorales por México-Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCyT), Mexico City, Mexico
| | - Angélica Serrano-Vázquez
- Laboratorio de Inmunología, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Héctor Godínez-Alvarez
- Unidad de Biotecnología y Prototipos, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City, Mexico State, Mexico
| | - Enrique González
- Laboratorio de Inmunología, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Liliana Rojas-Velázquez
- Laboratorio de Inmunología, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Patricia Moran
- Laboratorio de Inmunología, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - Manuel Ramiro
- División de Estudios de Posgrado, Facultad de Medicina, Universidad Nacional Autónoma de Mexico, Mexico City, Mexico
| | - Eric Hernández
- Laboratorio de Inmunología, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Clara Lau
- Laboratorios de Análisis Clínicos e Imagenología, Biomédica de Referencia, S.A.P.I. DE C.V., Mexico City, Mexico
| | - Marcela Martínez
- Laboratorios de Análisis Clínicos e Imagenología, Biomédica de Referencia, S.A.P.I. DE C.V., Mexico City, Mexico
| | - Ma. de los Ángeles Padilla
- Laboratorio de Inmunología, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Martha E. Zaragoza
- Laboratorio de Inmunología, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Blanca Taboada
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca Morelos, Mexico
| | - Laura A. Palomares
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca Morelos, Mexico
| | - Susana López
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca Morelos, Mexico
| | - Alejandro Alagón
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca Morelos, Mexico
| | - Carlos F. Arias
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca Morelos, Mexico
| | - Cecilia Ximénez
- Laboratorio de Inmunología, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
64
|
Ahluwalia P, Vashisht A, Singh H, Sahajpal NS, Mondal AK, Jones K, Farmaha J, Bloomquist R, Carlock CM, Fransoso D, Sun C, Day T, Prah C, Vuong T, Ray P, Bradshaw D, Galvis MM, Fulzele S, Raval G, Moore JX, Cortes J, James JN, Kota V, Kolhe R. Ethno-demographic disparities in humoral responses to the COVID-19 vaccine among healthcare workers. J Med Virol 2023; 95:e29067. [PMID: 37675796 PMCID: PMC10536788 DOI: 10.1002/jmv.29067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/14/2023] [Accepted: 08/23/2023] [Indexed: 09/08/2023]
Abstract
The COVID-19 pandemic had a profound impact on global health, but rapid vaccine administration resulted in a significant decline in morbidity and mortality rates worldwide. In this study, we sought to explore the temporal changes in the humoral immune response against SARS-CoV-2 healthcare workers (HCWs) in Augusta, GA, USA, and investigate any potential associations with ethno-demographic features. Specifically, we aimed to compare the naturally infected individuals with naïve individuals to understand the immune response dynamics after SARS-CoV-2 vaccination. A total of 290 HCWs were included and assessed prospectively in this study. COVID status was determined using a saliva-based COVID assay. Neutralizing antibody (NAb) levels were quantified using a chemiluminescent immunoassay system, and IgG levels were measured using an enzyme-linked immunosorbent assay method. We examined the changes in antibody levels among participants using different statistical tests including logistic regression and multiple correspondence analysis. Our findings revealed a significant decline in NAb and IgG levels at 8-12 months postvaccination. Furthermore, a multivariable analysis indicated that this decline was more pronounced in White HCWs (odds ratio [OR] = 2.1, 95% confidence interval [CI] = 1.07-4.08, p = 0.02) and IgG (OR = 2.07, 95% CI = 1.04-4.11, p = 0.03) among the whole cohort. Booster doses significantly increased IgG and NAb levels, while a decline in antibody levels was observed in participants without booster doses at 12 months postvaccination. Our results highlight the importance of understanding the dynamics of immune response and the potential influence of demographic factors on waning immunity to SARS-CoV-2. In addition, our findings emphasize the value of booster doses to ensure durable immunity.
Collapse
Affiliation(s)
- Pankaj Ahluwalia
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Ashutosh Vashisht
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Harmanpreet Singh
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | | | - Ashis K. Mondal
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Kimya Jones
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Jaspreet Farmaha
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
- Dental College of Georgia, Augusta University, GA, U.S.A
| | | | | | - Drew Fransoso
- Dental College of Georgia, Augusta University, GA, U.S.A
| | - Christina Sun
- Dental College of Georgia, Augusta University, GA, U.S.A
| | - Tyler Day
- Dental College of Georgia, Augusta University, GA, U.S.A
| | - Comfort Prah
- Dental College of Georgia, Augusta University, GA, U.S.A
| | - Trinh Vuong
- Dental College of Georgia, Augusta University, GA, U.S.A
| | - Patty Ray
- Clinical Trials Office, Augusta University, GA, U.S.A
| | | | | | - Sadanand Fulzele
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Girindra Raval
- Georgia Cancer Center at Augusta University, Augusta, GA 30912, USA
| | | | - Jorge Cortes
- Georgia Cancer Center at Augusta University, Augusta, GA 30912, USA
| | | | - Vamsi Kota
- Georgia Cancer Center at Augusta University, Augusta, GA 30912, USA
| | - Ravindra Kolhe
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
65
|
Singh G, Abbad A, Tcheou J, Mendu DR, Firpo-Betancourt A, Gleason C, Srivastava K, Cordon-Cardo C, Simon V, Krammer F, Carreño JM. Binding and Avidity Signatures of Polyclonal Sera From Individuals With Different Exposure Histories to Severe Acute Respiratory Syndrome Coronavirus 2 Infection, Vaccination, and Omicron Breakthrough Infections. J Infect Dis 2023; 228:564-575. [PMID: 37104046 PMCID: PMC10469125 DOI: 10.1093/infdis/jiad116] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 04/07/2023] [Accepted: 04/18/2023] [Indexed: 04/28/2023] Open
Abstract
BACKGROUND The number of exposures to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and to vaccine antigens affect the magnitude and avidity of the polyclonal response. METHODS We studied binding and avidity of different antibody isotypes to the spike, the receptor-binding domain (RBD), and the nucleoprotein (NP) of wild-type (WT) and BA.1 SARS-CoV-2 in convalescent, mRNA vaccinated and/or boosted, hybrid immune individuals and in individuals with breakthrough cases during the peak of the BA.1 wave. RESULTS We found an increase in spike-binding antibodies and antibody avidity with increasing number of exposures to infection and/or vaccination. NP antibodies were detectible in convalescent individuals and a proportion of breakthrough cases, but they displayed low avidity. Omicron breakthrough infections elicited high levels of cross-reactive antibodies between WT and BA.1 antigens in vaccinated individuals without prior infection directed against the spike and RBD. The magnitude of the antibody response and avidity correlated with neutralizing activity against WT virus. CONCLUSIONS The magnitude and quality of the antibody response increased with the number of antigenic exposures, including breakthrough infections. However, cross-reactivity of the antibody response after BA.1 breakthroughs, was affected by the number of prior exposures.
Collapse
Affiliation(s)
- Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Anass Abbad
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Johnstone Tcheou
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Demodara Rao Mendu
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Adolfo Firpo-Betancourt
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Charles Gleason
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Komal Srivastava
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Carlos Cordon-Cardo
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Viviana Simon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
66
|
Zhang J, Cong Y, Duan L, Zhang JZH. Combined Antibodies Evusheld against the SARS-CoV-2 Omicron Variants BA.1.1 and BA.5: Immune Escape Mechanism from Molecular Simulation. J Chem Inf Model 2023; 63:5297-5308. [PMID: 37586058 DOI: 10.1021/acs.jcim.3c00813] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
The Omicron lineage of SARS-CoV-2, which was first reported in November 2021, has spread globally and become dominant, splitting into several sublineages. Experiments have shown that Omicron lineage has escaped or reduced the activity of existing monoclonal antibodies, but the origin of escape mechanism caused by mutation is still unknown. This work uses molecular dynamics and umbrella sampling methods to reveal the escape mechanism of BA.1.1 to monoclonal antibody (mAb) Tixagevimab (AZD1061) and BA.5 to mAb Cilgavimab (AZD8895), both mAbs were combined to form antibody cocktail, Evusheld (AZD7442). The binding free energy of BA.1.1-AZD1061 and BA.5-AZD8895 has been severely reduced due to multiple-site mutated Omicron variants. Our results show that the two Omicron variants, which introduce a substantial number of positively charged residues, can weaken the electrostatic attraction between the receptor binding domain (RBD) and AZD7442, thus leading to a decrease in affinity. Additionally, using umbrella sampling along dissociation pathway, we found that the two Omicron variants severely impaired the interaction between the RBD of SARS-CoV-2's spike glycoprotein (S protein) and complementary determining regions (CDRs) of mAbs, especially in CDR3H. Although mAbs AZD8895 and AZD1061 are knocked out by BA.5 and BA.1.1, respectively, our results confirm that the antibody cocktail AZD7442 retains activity against BA.1.1 and BA.5 because another antibody is still on guard. The study provides theoretical insights for mAbs interacting with BA.1.1 and BA.5 from both energetic and dynamic perspectives, and we hope this will help in developing new monoclonals and combinations to protect those unable to mount adequate vaccine responses.
Collapse
Affiliation(s)
- Jianwen Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Shanghai Key Laboratory of Green Chemistry & Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Yalong Cong
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Shanghai Key Laboratory of Green Chemistry & Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Lili Duan
- School of Physics and Electronics, Shandong Normal University, Jinan 250014, China
| | - John Z H Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Shanghai Key Laboratory of Green Chemistry & Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
- Faculty of Synthetic Biology and Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- NYU-ECNU Center for Computational Chemistry at NYU Shanghai, Shanghai 200062, China
- Department of Chemistry, New York University, New York, New York 10003, United States
- Collaborative Innovation Center of Extreme Optics, Shanxi University, Taiyuan, Shanxi 030006, China
| |
Collapse
|
67
|
Al-Shudifat AE, Al-Tamimi M, Dawoud R, Alkhateeb M, Mryyian A, Alahmad A, Abbas MM, Qaqish A. Anti-S and Anti-N Antibody Responses of COVID-19 Vaccine Recipients. Vaccines (Basel) 2023; 11:1398. [PMID: 37766076 PMCID: PMC10537031 DOI: 10.3390/vaccines11091398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/01/2023] [Accepted: 08/12/2023] [Indexed: 09/29/2023] Open
Abstract
The long-term immunoglobulin responses of COVID-19 vaccinations is important to determine the efficacy of these vaccinations. This study aimed to investigate and compare the long-term immunoglobulin response of COVID-19 vaccination recipients, using anti-S IgG, anti-N IgG, and IgM titer levels. This study included 267 participants, comprising individuals who tested positive for COVID-19 through PCR testing (n = 125), and those who received the Pfizer (n = 133), Sinopharm (n = 112), AstraZeneca (n = 20), or Sputnik (n = 2) vaccines. Female participants comprised the largest share of this study (n = 147, 55.1%). This study found that most participants had positive IgG antibodies, with 96.3% having anti-S IgG and 75.7% having anti-N IgG. Most participants (90.3%) tested negative for anti-N IgM antibodies. Sinopharm-vaccinated individuals exhibited a notably lower rate of positive anti-S IgG (93.8%) and a significantly higher rate of positive anti-N IgG antibodies (91%). Anti-N IgG levels were significantly correlated with the number of prior COVID-19 infections (p = 0.015). Specifically, individuals with a history of four COVID-19 infections had higher anti-N IgG titers (14.1 ± 1.4) than those with only one experience of COVID-19 infection (9.4 ± 7.2). Individuals who were infected with COVID-19 after receiving the vaccine demonstrated higher levels of anti-N IgG, exhibiting a 25% increase in mean titer levels compared to those who were infected prior to vaccination. There was a statistically significant association between anti-N IgG positivity with age (p = 0.034), and smoking status (p = 0.006) of participants. Participants younger than 20 and older than 60 showed the highest positivity rate of anti-N (>90%). Smokers had a low positivity rate of anti-N (68.8%) compared to nonsmokers (83.6%). In conclusion, this study demonstrated that most COVID-19 vaccination recipients had positive IgG antibodies, with differences in the long-term immunoglobulin response depending on the type of vaccine administered and occurrence of COVID-19 infection.
Collapse
Affiliation(s)
- Abdel-Ellah Al-Shudifat
- Department of Internal and Family Medicine, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan;
| | - Mohammad Al-Tamimi
- Department of Microbiology, Pathology and Forensic Medicine, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan;
| | - Rand Dawoud
- Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan; (R.D.); (M.A.); (A.M.); (A.A.)
| | - Mohammad Alkhateeb
- Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan; (R.D.); (M.A.); (A.M.); (A.A.)
- Department of Internal Medicine, King Hussein Cancer Center, Amman 11941, Jordan
| | - Amel Mryyian
- Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan; (R.D.); (M.A.); (A.M.); (A.A.)
| | - Anas Alahmad
- Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan; (R.D.); (M.A.); (A.M.); (A.A.)
- Department of Internal Medicine, King Hussein Cancer Center, Amman 11941, Jordan
| | - Manal M Abbas
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan;
- Pharmacological and Diagnostic Research Lab, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Arwa Qaqish
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa 13133, Jordan
- Department of Cellular Therapy and Applied Genomics, King Hussein Cancer Center, Amman 11941, Jordan
| |
Collapse
|
68
|
Kanis FM, Meier JP, Guldan H, Niller HH, Dahm M, Dansard A, Zander T, Struck F, Soutschek E, Deml L, Möbus S, Barabas S. Performance of T-Track ® SARS-CoV-2, an Innovative Dual Marker RT-qPCR-Based Whole-Blood Assay for the Detection of SARS-CoV-2-Reactive T Cells. Diagnostics (Basel) 2023; 13:2722. [PMID: 37685260 PMCID: PMC10486492 DOI: 10.3390/diagnostics13172722] [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: 06/28/2023] [Revised: 08/07/2023] [Accepted: 08/11/2023] [Indexed: 09/10/2023] Open
Abstract
T-cell immunity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) plays a central role in the control of the virus. In this study, we evaluated the performance of T-Track® SARS-CoV-2, a novel CE-marked quantitative reverse transcription-polymerase chain reaction (RT-qPCR) assay, which relies on the combined evaluation of IFNG and CXCL10 mRNA levels in response to the S1 and NP SARS-CoV-2 antigens, in 335 participants with or without a history of SARS-CoV-2 infection and vaccination, respectively. Of the 62 convalescent donors, 100% responded to S1 and 88.7% to NP antigens. In comparison, of the 68 naïve donors, 4.4% were reactive to S1 and 19.1% to NP. Convalescent donors <50 and ≥50 years of age demonstrated a 100% S1 reactivity and an 89.1% and 87.5% NP reactivity, respectively. T-cell responses by T-Track® SARS-CoV-2 and IgG serology by recomLine SARS-CoV-2 IgG according to the time from the last immunisation (by vaccination or viral infection) were comparable. Both assays showed a persistent cellular and humoral response for at least 36 weeks post immunisation in vaccinated and convalescent donors. Our results demonstrate the very good performance of the T-Track® SARS-CoV-2 molecular assay and suggest that it might be suitable to monitor the SARS-CoV-2-specific T-cell response in COVID-19 vaccinations trials and cross-reactivity studies.
Collapse
Affiliation(s)
| | | | | | - Hans-Helmut Niller
- Institute for Medical Microbiology and Hygiene, University of Regensburg, 93053 Regensburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Aiello A, Najafi-Fard S, Goletti D. Initial immune response after exposure to Mycobacterium tuberculosis or to SARS-COV-2: similarities and differences. Front Immunol 2023; 14:1244556. [PMID: 37662901 PMCID: PMC10470049 DOI: 10.3389/fimmu.2023.1244556] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) and Coronavirus disease-2019 (COVID-19), whose etiologic agent is severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), are currently the two deadliest infectious diseases in humans, which together have caused about more than 11 million deaths worldwide in the past 3 years. TB and COVID-19 share several aspects including the droplet- and aerosol-borne transmissibility, the lungs as primary target, some symptoms, and diagnostic tools. However, these two infectious diseases differ in other aspects as their incubation period, immune cells involved, persistence and the immunopathological response. In this review, we highlight the similarities and differences between TB and COVID-19 focusing on the innate and adaptive immune response induced after the exposure to Mtb and SARS-CoV-2 and the pathological pathways linking the two infections. Moreover, we provide a brief overview of the immune response in case of TB-COVID-19 co-infection highlighting the similarities and differences of each individual infection. A comprehensive understanding of the immune response involved in TB and COVID-19 is of utmost importance for the design of effective therapeutic strategies and vaccines for both diseases.
Collapse
Affiliation(s)
| | | | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
70
|
Barrios MH, Nicholson S, Bull RA, Martinello M, Rawlinson W, Mina M, Post JJ, Hudson B, Gilroy N, Lloyd AR, Konecny P, Mordant F, Catton M, Subbarao K, Caly L, Druce J, Netter HJ. Comparative Longitudinal Serological Study of Anti-SARS-CoV-2 Antibody Profiles in People with COVID-19. Microorganisms 2023; 11:1985. [PMID: 37630545 PMCID: PMC10458948 DOI: 10.3390/microorganisms11081985] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/10/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
Serological diagnostic assays are essential tools for determining an individual's protection against viruses like SARS-CoV-2, tracking the spread of the virus in the community, and evaluating population immunity. To assess the diversity and quality of the anti-SARS-CoV-2 antibody response, we have compared the antibody profiles of people with mild, moderate, and severe COVID-19 using a dot blot assay. The test targeted the four major structural proteins of SARS-CoV-2, namely the nucleocapsid (N), spike (S) protein domains S1 and S2, and receptor-binding domain (RBD). Serum samples were collected from 63 participants at various time points for up to 300 days after disease onset. The dot blot assay revealed patient-specific differences in the anti-SARS-CoV-2 antibody profiles. Out of the 63 participants with confirmed SARS-CoV-2 infections and clinical COVID-19, 35/63 participants exhibited diverse and robust responses against the tested antigens, while 14/63 participants displayed either limited responses to a subset of antigens or no detectable antibody response to any of the antigens. Anti-N-specific antibody levels decreased within 300 days after disease onset, whereas anti-S-specific antibodies persisted. The dynamics of the antibody response did not change during the test period, indicating stable antibody profiles. Among the participants, 28/63 patients with restricted anti-S antibody profiles or undetectable anti-S antibody levels in the dot blot assay also exhibited weak neutralization activity, as measured by a surrogate virus neutralization test (sVNT) and a microneutralization test. These results indicate that in some cases, natural infections do not lead to the production of neutralizing antibodies. Furthermore, the study revealed significant serological variability among patients, regardless of the severity of their COVID-19 illness. These differences need to be carefully considered when evaluating the protective antibody status of individuals who have experienced primary SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Marilou H. Barrios
- Victorian Infectious Diseases Reference Laboratory (VIDRL), The Royal Melbourne Hospital, Melbourne, VIC 3000, Australia; (M.H.B.); (S.N.); (M.C.); (L.C.); (J.D.)
- Peter Doherty Institute, University of Melbourne, Melbourne, VIC 3000, Australia; (F.M.); (K.S.)
| | - Suellen Nicholson
- Victorian Infectious Diseases Reference Laboratory (VIDRL), The Royal Melbourne Hospital, Melbourne, VIC 3000, Australia; (M.H.B.); (S.N.); (M.C.); (L.C.); (J.D.)
- Peter Doherty Institute, University of Melbourne, Melbourne, VIC 3000, Australia; (F.M.); (K.S.)
| | - Rowena A. Bull
- The Kirby Institute, University of New South Wales (UNSW), Sydney, NSW 2052, Australia; (R.A.B.); (M.M.); (A.R.L.)
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales (UNSW), Sydney, NSW 2052, Australia;
| | - Marianne Martinello
- The Kirby Institute, University of New South Wales (UNSW), Sydney, NSW 2052, Australia; (R.A.B.); (M.M.); (A.R.L.)
| | - William Rawlinson
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales (UNSW), Sydney, NSW 2052, Australia;
- Serology and Virology Division, Department of Microbiology, New South Wales Health Pathology, Randwick, Sydney, NSW 2031, Australia
- Prince of Wales Hospital, Sydney, NSW 2031, Australia;
| | - Michael Mina
- Northern Beaches Hospital, Frenchs Forest, NSW 2086, Australia;
| | - Jeffrey J. Post
- Prince of Wales Hospital, Sydney, NSW 2031, Australia;
- School of Clinical Medicine, University of New South Wales (UNSW), Sydney, NSW 2052, Australia;
| | - Bernard Hudson
- Royal North Shore Hospital, Sydney, NSW 2065, Australia;
| | | | - Andrew R. Lloyd
- The Kirby Institute, University of New South Wales (UNSW), Sydney, NSW 2052, Australia; (R.A.B.); (M.M.); (A.R.L.)
| | - Pamela Konecny
- School of Clinical Medicine, University of New South Wales (UNSW), Sydney, NSW 2052, Australia;
- St. George Hospital, Sydney, NSW 2217, Australia
| | - Francesca Mordant
- Peter Doherty Institute, University of Melbourne, Melbourne, VIC 3000, Australia; (F.M.); (K.S.)
- Department of Microbiology and Immunology, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Mike Catton
- Victorian Infectious Diseases Reference Laboratory (VIDRL), The Royal Melbourne Hospital, Melbourne, VIC 3000, Australia; (M.H.B.); (S.N.); (M.C.); (L.C.); (J.D.)
- Peter Doherty Institute, University of Melbourne, Melbourne, VIC 3000, Australia; (F.M.); (K.S.)
| | - Kanta Subbarao
- Peter Doherty Institute, University of Melbourne, Melbourne, VIC 3000, Australia; (F.M.); (K.S.)
- Department of Microbiology and Immunology, University of Melbourne, Melbourne, VIC 3000, Australia
- World Health Organization Collaborating Centre for Reference and Research on Influenza at the Peter Doherty Institute, Melbourne, VIC 3000, Australia
| | - Leon Caly
- Victorian Infectious Diseases Reference Laboratory (VIDRL), The Royal Melbourne Hospital, Melbourne, VIC 3000, Australia; (M.H.B.); (S.N.); (M.C.); (L.C.); (J.D.)
- Peter Doherty Institute, University of Melbourne, Melbourne, VIC 3000, Australia; (F.M.); (K.S.)
| | - Julian Druce
- Victorian Infectious Diseases Reference Laboratory (VIDRL), The Royal Melbourne Hospital, Melbourne, VIC 3000, Australia; (M.H.B.); (S.N.); (M.C.); (L.C.); (J.D.)
- Peter Doherty Institute, University of Melbourne, Melbourne, VIC 3000, Australia; (F.M.); (K.S.)
| | - Hans J. Netter
- Victorian Infectious Diseases Reference Laboratory (VIDRL), The Royal Melbourne Hospital, Melbourne, VIC 3000, Australia; (M.H.B.); (S.N.); (M.C.); (L.C.); (J.D.)
- Peter Doherty Institute, University of Melbourne, Melbourne, VIC 3000, Australia; (F.M.); (K.S.)
- School of Science, Royal Melbourne Institute of Technology (RMIT) University, Melbourne, VIC 3001, Australia
| |
Collapse
|
71
|
Dias Assis BR, Gomes IP, de Castro JT, Rivelli GG, de Castro NS, Gomez-Mendoza DP, Bagno FF, Hojo-Souza NS, Chaves Maia AL, Lages EB, da Fonseca FG, Ribeiro Teixeira SM, Fernandes AP, Gazzinelli RT, Castro Goulart GA. Quality attributes of CTVad1, a nanoemulsified adjuvant for phase I clinical trial of SpiN COVID-19 vaccine. Nanomedicine (Lond) 2023; 18:1175-1194. [PMID: 37712604 DOI: 10.2217/nnm-2023-0122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023] Open
Abstract
Aim: To develop, characterize and evaluate an oil/water nanoemulsion with squalene (CTVad1) to be approved as an adjuvant for the SpiN COVID-19 vaccine clinical trials. Materials & methods: Critical process parameters (CPPs) of CTVad1 were standardized to meet the critical quality attributes (CQAs) of an adjuvant for human use. CTVad1 and the SpiN-CTVad1 vaccine were submitted to physicochemical, stability, in vitro and in vivo studies. Results & conclusion: All CQAs were met in the CTVad1 production process. SpiN- CTVad1 met CQAs and induced high levels of antibodies and specific cellular responses in in vivo studies. These results represented a critical step in the process developed to meet regulatory requirements for the SpiN COVID-19 vaccine clinical trial.
Collapse
Affiliation(s)
- Bruna Rodrigues Dias Assis
- Department of Pharmaceuticals, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Isabela Pereira Gomes
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Júlia Teixeira de Castro
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Graziella Gomes Rivelli
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Natália Salazar de Castro
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Diana Paola Gomez-Mendoza
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Flávia Fonseca Bagno
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Natália Satchiko Hojo-Souza
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Belo Horizonte, MG, 30190-002, Brazil
| | - Ana Luiza Chaves Maia
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Eduardo Burgarelli Lages
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Flávio Guimaraes da Fonseca
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Santuza Maria Ribeiro Teixeira
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Department of Biochemistry & Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Ana Paula Fernandes
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Department of Clinical & Toxicological Analysis, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Ricardo Tostes Gazzinelli
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Belo Horizonte, MG, 30190-002, Brazil
- Department of Biochemistry & Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Gisele Assis Castro Goulart
- Department of Pharmaceuticals, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| |
Collapse
|
72
|
Kumari S, Jeseena K, Kiran AK, Kujur M, Saroj U, Hembrom SS, Kujur A. Immunological survey of COVID-19 among medicos of tribal preponderant state of India. J Family Med Prim Care 2023; 12:1669-1672. [PMID: 37767453 PMCID: PMC10521841 DOI: 10.4103/jfmpc.jfmpc_272_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 09/29/2023] Open
Abstract
Background Immunological Survey or serosurveys have yielded useful information regarding the spread of the COVID-19 pandemic in the general population, but the impact of the continuing pandemic on the medical students in India is yet to be fully recognised. In this study we assessed the students who had received at least two doses of the COVID-19 vaccine for their antibody response. Methodology A Hospital based, age-stratified, cross-sectional Analytical study design was adopted for the survey, carried out in tribal state of India among medical students. Consecutive sampling method was used where serum samples were tested for antibodies against the SARS-CoV-2 nucleocapsid (N) protein. Result The vaccinee group comprised of 187 students mostly aged between 18-23 years 68.4% were females, 56.6 % were vaccinated with covishield. The mean IgG (Immunoglobin G) titre was 7343.74 AU/Ml, less than 1000 AU/Ml was found in 8% of participants, while more than 8000 AU/Ml was found in 32.1%. Participants who got the covaxin vaccine had a higher median IgG titre (median 6491.8 AU/mL, interquartile range 8898 AU/mL).The antibody titre of male was 0.328 times lower than that of female. Conclusion Despite the fact that covishield's mean antibody titre was higher, covaxin's protection lasted longer.
Collapse
Affiliation(s)
- Sushma Kumari
- Department of Blood Bank, Rajendra Institute of Medical Sciences (RIMS), Ranchi, Jharkhand, India
| | - K Jeseena
- Department of Community Medicine, Rajendra Institute of Medical Sciences (RIMS), Ranchi, Jharkhand, India
| | - Asha K. Kiran
- Department of Community Medicine, Rajendra Institute of Medical Sciences (RIMS), Ranchi, Jharkhand, India
| | - Manisha Kujur
- Department of Community Medicine, Rajendra Institute of Medical Sciences (RIMS), Ranchi, Jharkhand, India
| | - Usha Saroj
- Department of Blood Bank, Rajendra Institute of Medical Sciences (RIMS), Ranchi, Jharkhand, India
| | - Shailesh S. Hembrom
- Department of Community Medicine, Rajendra Institute of Medical Sciences (RIMS), Ranchi, Jharkhand, India
| | - Anit Kujur
- Department of Community Medicine, Rajendra Institute of Medical Sciences (RIMS), Ranchi, Jharkhand, India
| |
Collapse
|
73
|
Dukes CW, Rossetti RAM, Hensel JA, Snedal S, Cubitt CL, Schell MJ, Abrahamsen M, Isaacs-Soriano K, Kennedy K, Mangual LN, Whiting J, Martinez-Brockhus V, Islam JY, Rathwell J, Beatty M, Hall AM, Abate-Daga D, Giuliano AR, Pilon-Thomas S. SARS-CoV-2 antibody response duration and neutralization following natural infection. JOURNAL OF CLINICAL VIROLOGY PLUS 2023; 3:100158. [PMID: 37654784 PMCID: PMC10470471 DOI: 10.1016/j.jcvp.2023.100158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023] Open
Abstract
Background The role of the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) neutralizing antibody response from natural infection and vaccination, and the potential determinants of this response are poorly understood. Characterizing this antibody response and the factors associated with neutralization can help inform future prevention efforts and improve clinical outcomes in those infected. Objectives The goals of this study were to prospectively evaluate SARS-CoV-2 antibody levels and the neutralizing antibody responses among naturally infected adults and to determine demographic and behavioral factors independently associated with these responses. Methods Serum was collected from seropositive individuals at baseline, four-weeks, and three-months following their first study visit to be evaluated for antibody levels. Detection of neutralizing antibodies was performed at baseline. Participant demographic and behavioral information was collected via web questionnaire prior to their first visit. Results At baseline, higher antibody levels were associated with better neutralization capacity, with 83% of participants having detectable neutralizing antibodies. We found an age-dependent effect on antibody level and neutralization capacity with participants over 65 years having significantly higher levels. Ethnicity, heart disease, autoimmune disease, and COVID symptoms were associated with higher antibody levels, but not with increased neutralization capacity. Work environment during the pandemic correlated with increased neutralization capacity, while kidney or liver disease and traveling out of state after February 2020 correlated with decreased neutralization capacity, however neither correlated with antibody levels. Conclusions Our data show that natural infection by SARS-CoV-2 can induce a humoral response reflected by high antibody levels and neutralization capacity.
Collapse
Affiliation(s)
- Christopher W Dukes
- Department of Immunology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, United States of America
- Center for Immunization and Infection Research in Cancer, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida, 33612, United States of America
| | - Renata AM Rossetti
- Department of Immunology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, United States of America
| | - Jonathan A Hensel
- Department of Immunology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, United States of America
| | - Sebastian Snedal
- Department of Immunology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, United States of America
| | - Christopher L Cubitt
- Immune Monitoring Core Facility, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida, 33612, United States of America
| | - Michael J Schell
- Center for Immunization and Infection Research in Cancer, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida, 33612, United States of America
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida, 33612, United States of America
| | - Martha Abrahamsen
- Department of Cancer Epidemiology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida, 33612, United States of America
| | - Kimberly Isaacs-Soriano
- Department of Cancer Epidemiology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida, 33612, United States of America
| | - Kayoko Kennedy
- Department of Cancer Epidemiology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida, 33612, United States of America
| | - Leslie N Mangual
- Department of Cancer Epidemiology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida, 33612, United States of America
| | - Junmin Whiting
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida, 33612, United States of America
| | - Veronica Martinez-Brockhus
- Department of Immunology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, United States of America
| | - Jessica Y Islam
- Center for Immunization and Infection Research in Cancer, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida, 33612, United States of America
- Department of Cancer Epidemiology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida, 33612, United States of America
| | - Julie Rathwell
- Center for Immunization and Infection Research in Cancer, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida, 33612, United States of America
- Department of Cancer Epidemiology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida, 33612, United States of America
| | - Matthew Beatty
- Department of Immunology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, United States of America
| | - Amy M Hall
- Department of Immunology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, United States of America
| | - Daniel Abate-Daga
- Department of Immunology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, United States of America
- Center for Immunization and Infection Research in Cancer, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida, 33612, United States of America
| | - Anna R Giuliano
- Center for Immunization and Infection Research in Cancer, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida, 33612, United States of America
- Department of Cancer Epidemiology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida, 33612, United States of America
| | - Shari Pilon-Thomas
- Department of Immunology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, United States of America
- Center for Immunization and Infection Research in Cancer, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida, 33612, United States of America
| |
Collapse
|
74
|
Martel F, Cuervo-Rojas J, Ángel J, Ariza B, González JM, Ramírez-Santana C, Acosta-Ampudia Y, Murcia-Soriano L, Montoya N, Cardozo-Romero CC, Valderrama-Beltrán SL, Cepeda M, Castellanos JC, Gómez-Restrepo C, Perdomo-Celis F, Gazquez A, Dickson A, Brien JD, Mateus J, Grifoni A, Sette A, Weiskopf D, Franco MA. Cross-reactive humoral and CD4 + T cell responses to Mu and Gamma SARS-CoV-2 variants in a Colombian population. Front Immunol 2023; 14:1241038. [PMID: 37575243 PMCID: PMC10413264 DOI: 10.3389/fimmu.2023.1241038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 07/07/2023] [Indexed: 08/15/2023] Open
Abstract
The SARS CoV-2 antibody and CD4+ T cell responses induced by natural infection and/or vaccination decline over time and cross-recognize other viral variants at different levels. However, there are few studies evaluating the levels and durability of the SARS CoV-2-specific antibody and CD4+ T cell response against the Mu, Gamma, and Delta variants. Here, we examined, in two ambispective cohorts of naturally-infected and/or vaccinated individuals, the titers of anti-RBD antibodies and the frequency of SARS-CoV-2-specific CD4+ T cells up to 6 months after the last antigen exposure. In naturally-infected individuals, the SARS-CoV-2 antibody response declined 6 months post-symptoms onset. However, the kinetic observed depended on the severity of the disease, since individuals who developed severe COVID-19 maintained the binding antibody titers. Also, there was detectable binding antibody cross-recognition for the Gamma, Mu, and Delta variants, but antibodies poorly neutralized Mu. COVID-19 vaccines induced an increase in antibody titers 15-30 days after receiving the second dose, but these levels decreased at 6 months. However, as expected, a third dose of the vaccine caused a rise in antibody titers. The dynamics of the antibody response upon vaccination depended on the previous SARS-CoV-2 exposure. Lower levels of vaccine-induced antibodies were associated with the development of breakthrough infections. Vaccination resulted in central memory spike-specific CD4+ T cell responses that cross-recognized peptides from the Gamma and Mu variants, and their duration also depended on previous SARS-CoV-2 exposure. In addition, we found cross-reactive CD4+ T cell responses in unexposed and unvaccinated individuals. These results have important implications for vaccine design for new SARS-CoV-2 variants of interest and concern.
Collapse
Affiliation(s)
- Fabiola Martel
- Institute of Human Genetics, School of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Juliana Cuervo-Rojas
- Department of Clinical Epidemiology and Biostatistics, School of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Juana Ángel
- Institute of Human Genetics, School of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Beatriz Ariza
- Clinical Laboratory Science Research Group, Clinical Laboratory, Hospital Universitario San Ignacio, Bogotá, Colombia
| | - John Mario González
- Group of Basic Medical Sciences, School of Medicine, Universidad de Los Andes, Bogotá, Colombia
| | - Carolina Ramírez-Santana
- Center for Autoimmune Diseases Research
(CREA), School of Medicine and Health Sciences, Universidad del Rosario,, Bogotá, Colombia
| | - Yeny Acosta-Ampudia
- Center for Autoimmune Diseases Research
(CREA), School of Medicine and Health Sciences, Universidad del Rosario,, Bogotá, Colombia
| | | | - Norma Montoya
- Head Clinical Laboratory Unit, Clínica del Occidente, Bogotá, Colombia
| | | | - Sandra Liliana Valderrama-Beltrán
- Division of Infectious Diseases, Department of Internal Medicine. School of Medicine, Pontificia Universidad Javeriana, Hospital Universitario San Ignacio Infectious Diseases Research Group, Bogotá, Colombia
| | - Magda Cepeda
- Department of Clinical Epidemiology and Biostatistics, School of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | - Carlos Gómez-Restrepo
- Department of Clinical Epidemiology and Biostatistics, School of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Federico Perdomo-Celis
- Institute of Human Genetics, School of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Andreu Gazquez
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Alexandria Dickson
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - James D. Brien
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - José Mateus
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA, United States
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Manuel A. Franco
- Institute of Human Genetics, School of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
75
|
Bansal A, Trieu MC, Mohn KGI, Madsen A, Olofsson JS, Sandnes HH, Sævik M, Søyland H, Hansen L, Onyango TB, Tøndel C, Brokstad KA, Syre H, Riis ÅG, Langeland N, Cox RJ. Risk assessment and antibody responses to SARS-CoV-2 in healthcare workers. Front Public Health 2023; 11:1164326. [PMID: 37546332 PMCID: PMC10402899 DOI: 10.3389/fpubh.2023.1164326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 06/22/2023] [Indexed: 08/08/2023] Open
Abstract
Background Preventing infection in healthcare workers (HCWs) is crucial for protecting healthcare systems during the COVID-19 pandemic. Here, we investigated the seroepidemiology of SARS-CoV-2 in HCWs in Norway with low-transmission settings. Methods From March 2020, we recruited HCWs at four medical centres. We determined infection by SARS-CoV-2 RT-PCR and serological testing and evaluated the association between infection and exposure variables, comparing our findings with global data in a meta-analysis. Anti-spike IgG antibodies were measured after infection and/or vaccination in a longitudinal cohort until June 2021. Results We identified a prevalence of 10.5% (95% confidence interval, CI: 8.8-12.3) in 2020 and an incidence rate of 15.0 cases per 100 person-years (95% CI: 12.5-17.8) among 1,214 HCWs with 848 person-years of follow-up time. Following infection, HCWs (n = 63) mounted durable anti-spike IgG antibodies with a half-life of 4.3 months since their seropositivity. HCWs infected with SARS-CoV-2 in 2020 (n = 46) had higher anti-spike IgG titres than naive HCWs (n = 186) throughout the 5 months after vaccination with BNT162b2 and/or ChAdOx1-S COVID-19 vaccines in 2021. In a meta-analysis including 20 studies, the odds ratio (OR) for SARS-CoV-2 seropositivity was significantly higher with household contact (OR 12.6; 95% CI: 4.5-35.1) and occupational exposure (OR 2.2; 95% CI: 1.4-3.2). Conclusion We found high and modest risks of SARS-CoV-2 infection with household and occupational exposure, respectively, in HCWs, suggesting the need to strengthen infection prevention strategies within households and medical centres. Infection generated long-lasting antibodies in most HCWs; therefore, we support delaying COVID-19 vaccination in primed HCWs, prioritising the non-infected high-risk HCWs amid vaccine shortage.
Collapse
Affiliation(s)
- Amit Bansal
- Department of Clinical Science, Influenza Centre, University of Bergen, Bergen, Norway
| | - Mai-Chi Trieu
- Department of Clinical Science, Influenza Centre, University of Bergen, Bergen, Norway
| | - Kristin G. I. Mohn
- Department of Clinical Science, Influenza Centre, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Anders Madsen
- Department of Clinical Science, Influenza Centre, University of Bergen, Bergen, Norway
| | - Jan Stefan Olofsson
- Department of Clinical Science, Influenza Centre, University of Bergen, Bergen, Norway
| | | | - Marianne Sævik
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Hanne Søyland
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Lena Hansen
- Department of Clinical Science, Influenza Centre, University of Bergen, Bergen, Norway
| | | | - Camilla Tøndel
- Department of Clinical Science, Influenza Centre, University of Bergen, Bergen, Norway
- Department of Paediatrics, Haukeland University Hospital, Bergen, Norway
| | - Karl Albert Brokstad
- Department of Clinical Science, Influenza Centre, University of Bergen, Bergen, Norway
- Department of Safety, Chemistry and Biomedical Laboratory Sciences, Western Norway University of Applied Sciences, Bergen, Norway
| | | | - Heidi Syre
- Department of Medical Microbiology, Stavanger University Hospital, Stavanger, Norway
| | - Åse Garløv Riis
- Department of Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Nina Langeland
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Rebecca Jane Cox
- Department of Clinical Science, Influenza Centre, University of Bergen, Bergen, Norway
- Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
76
|
Bang MS, Kim CM, Cho NH, Seo JW, Kim DY, Yun NR, Kim DM. Evaluation of humoral immune response in relation to COVID-19 severity over 1 year post-infection: critical cases higher humoral immune response than mild cases. Front Immunol 2023; 14:1203803. [PMID: 37545518 PMCID: PMC10401267 DOI: 10.3389/fimmu.2023.1203803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/03/2023] [Indexed: 08/08/2023] Open
Abstract
Introduction Coronavirus disease 2019 (COVID-19) is caused by SARS-CoV-2. We investigated the antibody response against SARS-CoV-2 until 1 year after symptom onset. Methods We collected 314 serum samples from 97 patients with COVID-19. Antibody responses were tested using an indirect immunofluorescence assay (IFA), enzyme-linked immunosorbent assay (ELISA), and plaque reduction neutralization test (PRNT) to detect specific neutralizing antibodies. Results The positivity rates for neutralizing antibodies at a 1:10 titer cutoff were 58.1% at 1 week, 97.8% at 4 weeks, and 78% at 1 year after symptom onset (53.8% in asymptomatic patients and 89.3% in symptomatic patients). The IFA and anti-S1 ELISA IgG results significantly correlated with neutralizing antibody titers. Critical/fatal cases showed significantly higher antibody titers than the asymptomatic or mild-to-moderate illness groups. Nonetheless, the median number of days to the seroconversion of neutralizing antibodies was 10 and 15 in asymptomatic and symptomatic patients, respectively. The asymptomatic group had a significantly higher neutralizing potency index than the mild-to-severe illness groups. Conclusions Neutralizing antibodies corresponded to earlier seroconversion but had a shorter presence in the asymptomatic group than in the symptomatic group and were still present 1 year after symptom onset in critical/fatal cases.
Collapse
Affiliation(s)
- Mi-Seon Bang
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju, Republic of Korea
| | - Choon-Mee Kim
- Premedical Science, College of Medicine, Chosun University, Gwangju, Republic of Korea
| | - Nam-Hyuk Cho
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jun-Won Seo
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju, Republic of Korea
| | - Da Young Kim
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju, Republic of Korea
| | - Na Ra Yun
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju, Republic of Korea
| | - Dong-Min Kim
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju, Republic of Korea
| |
Collapse
|
77
|
Amellal H, Assaid N, Charoute H, Akarid K, Maaroufi A, Ezzikouri S, Sarih M. Kinetics of specific anti-SARS-CoV-2 IgM, IgA, and IgG responses during the first 12 months after SARS-CoV-2 infection: A prospective longitudinal study. PLoS One 2023; 18:e0288557. [PMID: 37437051 DOI: 10.1371/journal.pone.0288557] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 06/29/2023] [Indexed: 07/14/2023] Open
Abstract
Coronavirus 2019 (COVID-19) is a global health threat. The kinetics of antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) need to be assessed, as the long-term duration of these immunoglobulins remains largely controversial. The aim of this study was to assess the longitudinal dynamics of anti-SARS-CoV-2 antibodies against the nucleocapsid (N) protein and the receptor-binding domain (RBD) of the spike protein up to one year in a cohort of 190 COVID-19 patients. Between March and September 2021, we enrolled patients from two regional hospitals in Casablanca, Morocco. Blood samples were collected and analyzed for antibody levels. We used the commercial Euroimmun ELISA for the determination of anti-N IgM, the Abbott Architect™ SARS-CoV-2 IgG test for the detection of anti-RBD IgG, and an in-house kit for the assay of anti-N IgG and anti-N IgA. IgM and IgA antibodies were assessed 2-5, 9-12, 17-20 and 32-37 days after symptom onset. IgG antibodies were also assessed 60, 90, 120 and 360 days after symptom onset. One-third of patients developed IgM (32%), while two-thirds developed IgA (61%). One month of symptom onset, most patients developed IgG, with 97% and 93% positivity for anti-RBD IgG and anti-N IgG, respectively. The anti-RBD IgG positivity rate remained high up to one year of follow-up. However, the anti-N IgG positivity rate decreased over time, with only 41% of patients testing positive after one year's follow-up. IgG levels were significantly higher in older people (over 50 years) than in other study participants. We also found that patients who had received two doses of ChAdOx1 nCoV-19 vaccine prior to infection had a lower IgM response than unvaccinated patients. This difference was statistically significant two weeks after the onset of symptoms. We present the first study in Africa to measure the kinetics of antibody response (IgA, IgM and IgG) to SARS-CoV-2 over one year. Most participants remained seropositive for anti-RBD IgG after one year but showed a significant decline in antibody titers.
Collapse
Affiliation(s)
- Houda Amellal
- Department of Parasitology and Vector-Borne Diseases, Institut Pasteur du Maroc, Casablanca, Morocco
- Aïn Chock Faculty of Sciences, Health and Environment Laboratory, Biochemistry, Biotechnology and Immunophysiopathology Research Team, Hassan II University of Casablanca, Casablanca, Morocco
| | - Najlaa Assaid
- Department of Parasitology and Vector-Borne Diseases, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Hicham Charoute
- Institut Pasteur du Maroc, Research Unit of Epidemiology, Biostatistics and Bioinformatics, Casablanca, Morocco
| | - Khadija Akarid
- Aïn Chock Faculty of Sciences, Health and Environment Laboratory, Biochemistry, Biotechnology and Immunophysiopathology Research Team, Hassan II University of Casablanca, Casablanca, Morocco
| | - Abderrahmane Maaroufi
- Department of Parasitology and Vector-Borne Diseases, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Sayeh Ezzikouri
- Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Virology Unit, Casablanca, Morocco
| | - M'hammed Sarih
- Department of Parasitology and Vector-Borne Diseases, Institut Pasteur du Maroc, Casablanca, Morocco
| |
Collapse
|
78
|
Purcell RA, Theisen RM, Arnold KB, Chung AW, Selva KJ. Polyfunctional antibodies: a path towards precision vaccines for vulnerable populations. Front Immunol 2023; 14:1183727. [PMID: 37600816 PMCID: PMC10433199 DOI: 10.3389/fimmu.2023.1183727] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/30/2023] [Indexed: 08/22/2023] Open
Abstract
Vaccine efficacy determined within the controlled environment of a clinical trial is usually substantially greater than real-world vaccine effectiveness. Typically, this results from reduced protection of immunologically vulnerable populations, such as children, elderly individuals and people with chronic comorbidities. Consequently, these high-risk groups are frequently recommended tailored immunisation schedules to boost responses. In addition, diverse groups of healthy adults may also be variably protected by the same vaccine regimen. Current population-based vaccination strategies that consider basic clinical parameters offer a glimpse into what may be achievable if more nuanced aspects of the immune response are considered in vaccine design. To date, vaccine development has been largely empirical. However, next-generation approaches require more rational strategies. We foresee a generation of precision vaccines that consider the mechanistic basis of vaccine response variations associated with both immunogenetic and baseline health differences. Recent efforts have highlighted the importance of balanced and diverse extra-neutralising antibody functions for vaccine-induced protection. However, in immunologically vulnerable populations, significant modulation of polyfunctional antibody responses that mediate both neutralisation and effector functions has been observed. Here, we review the current understanding of key genetic and inflammatory modulators of antibody polyfunctionality that affect vaccination outcomes and consider how this knowledge may be harnessed to tailor vaccine design for improved public health.
Collapse
Affiliation(s)
- Ruth A. Purcell
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Robert M. Theisen
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Kelly B. Arnold
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Amy W. Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Kevin J. Selva
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
79
|
Petrone L, Sette A, de Vries RD, Goletti D. The Importance of Measuring SARS-CoV-2-Specific T-Cell Responses in an Ongoing Pandemic. Pathogens 2023; 12:862. [PMID: 37513709 PMCID: PMC10385870 DOI: 10.3390/pathogens12070862] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Neutralizing antibodies are considered a correlate of protection against SARS-CoV-2 infection and severe COVID-19, although they are not the only contributing factor to immunity: T-cell responses are considered important in protecting against severe COVID-19 and contributing to the success of vaccination effort. T-cell responses after vaccination largely mirror those of natural infection in magnitude and functional capacity, but not in breadth, as T-cells induced by vaccination exclusively target the surface spike glycoprotein. T-cell responses offer a long-lived line of defense and, unlike humoral responses, largely retain reactivity against the SARS-CoV-2 variants. Given the increasingly recognized role of T-cell responses in protection against severe COVID-19, the circulation of SARS-CoV-2 variants, and the potential implementation of novel vaccines, it becomes imperative to continuously monitor T-cell responses. In addition to "classical" T-cell assays requiring the isolation of peripheral blood mononuclear cells, simple whole-blood-based interferon-γ release assays have a potential role in routine T-cell response monitoring. These assays could be particularly useful for immunocompromised people and other clinically vulnerable populations, where interactions between cellular and humoral immunity are complex. As we continue to live alongside COVID-19, the importance of considering immunity as a whole, incorporating both humoral and cellular responses, is crucial.
Collapse
Affiliation(s)
- Linda Petrone
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS, 00149 Rome, Italy;
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA;
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Rory D. de Vries
- Department Viroscience, Erasmus University Medical Center, 3015CN Rotterdam, The Netherlands;
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS, 00149 Rome, Italy;
| |
Collapse
|
80
|
Huang C, Huang L, Wang Y, Li X, Ren L, Gu X, Kang L, Guo L, Liu M, Zhou X, Luo J, Huang Z, Tu S, Zhao Y, Chen L, Xu D, Li Y, Li C, Peng L, Li Y, Xie W, Cui D, Shang L, Fan G, Xu J, Wang G, Wang Y, Zhong J, Wang C, Wang J, Zhang D, Cao B. 6-month consequences of COVID-19 in patients discharged from hospital: a cohort study. Lancet 2023; 401:e21-e33. [PMID: 37321233 PMCID: PMC10258565 DOI: 10.1016/s0140-6736(23)00810-3] [Citation(s) in RCA: 107] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/02/2023] [Accepted: 04/13/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND The long-term health consequences of COVID-19 remain largely unclear. The aim of this study was to describe the long-term health consequences of patients with COVID-19 who have been discharged from hospital and investigate the associated risk factors, in particular disease severity. METHODS We did an ambidirectional cohort study of patients with confirmed COVID-19 who had been discharged from Jin Yin-tan Hospital (Wuhan, China) between Jan 7 and May 29, 2020. Patients who died before follow-up; patients for whom follow-up would be difficult because of psychotic disorders, dementia, or readmission to hospital; those who were unable to move freely due to concomitant osteoarthropathy or immobile before or after discharge due to diseases such as stroke or pulmonary embolism; those who declined to participate; those who could not be contacted; and those living outside of Wuhan or in nursing or welfare homes were all excluded. All patients were interviewed with a series of questionnaires for evaluation of symptoms and health-related quality of life, underwent physical examinations and a 6-min walking test, and received blood tests. A stratified sampling procedure was used to sample patients according to their highest seven-category scale during their hospital stay as 3, 4, and 5-6, to receive pulmonary function test, high resolution CT of the chest, and ultrasonography. Enrolled patients who had participated in the Lopinavir Trial for Suppression of SARS-CoV-2 in China received SARS-CoV-2 antibody tests. Multivariable adjusted linear or logistic regression models were used to evaluate the association between disease severity and long-term health consequences. FINDINGS In total, 1733 of 2469 discharged patients with COVID-19 were enrolled after 736 were excluded. Patients had a median age of 57·0 years (IQR 47·0-65·0) and 897 (52%) were male and 836 (48%) were female. The follow-up study was done from June 16 to Sept 3, 2020, and the median follow-up time after symptom onset was 186·0 days (175·0-199·0). Fatigue or muscle weakness (52%, 855 of 1654) and sleep difficulties (26%, 437 of 1655) were the most common symptoms. Anxiety or depression was reported among 23% (367 of 1616) of patients. The proportions of 6-min walking distance less than the lower limit of the normal range were 17% for those at severity scale 3, 13% for severity scale 4, and 28% for severity scale 5-6. The corresponding proportions of patients with diffusion impairment were 22% for severity scale 3, 29% for scale 4, and 56% for scale 5-6, and median CT scores were 3·0 (IQR 2·0-5·0) for severity scale 3, 4·0 (3·0-5·0) for scale 4, and 5·0 (4·0-6·0) for scale 5-6. After multivariable adjustment, patients showed an odds ratio (OR) of 1·61 (95% CI 0·80-3·25) for scale 4 versus scale 3 and 4·60 (1·85-11·48) for scale 5-6 versus scale 3 for diffusion impairment; OR 0·88 (0·66-1·17) for scale 4 versus scale 3 and OR 1·76 (1·05-2·96) for scale 5-6 versus scale 3 for anxiety or depression, and OR 0·87 (0·68-1·11) for scale 4 versus scale 3 and 2·75 (1·61-4·69) for scale 5-6 versus scale 3 for fatigue or muscle weakness. Of 94 patients with blood antibodies tested at follow-up, the seropositivity (96·2% vs 58·5%) and median titres (19·0 vs 10·0) of the neutralising antibodies were significantly lower compared with at the acute phase. 107 of 822 participants without acute kidney injury and with an estimated glomerular filtration rate (eGFR) of 90 mL/min per 1·73 m2 or more at acute phase had eGFR less than 90 mL/min per 1·73 m2 at follow-up. INTERPRETATION At 6 months after acute infection, COVID-19 survivors were mainly troubled with fatigue or muscle weakness, sleep difficulties, and anxiety or depression. Patients who were more severely ill during their hospital stay had more severe impaired pulmonary diffusion capacities and abnormal chest imaging manifestations, and are the main target population for intervention of long-term recovery. FUNDING National Natural Science Foundation of China, Chinese Academy of Medical Sciences Innovation Fund for Medical Sciences, National Key Research and Development Program of China, Major Projects of National Science and Technology on New Drug Creation and Development of Pulmonary Tuberculosis, and Peking Union Medical College Foundation.
Collapse
Affiliation(s)
- Chaolin Huang
- Medical Department, Jin Yin-tan Hospital, Wuhan, China; Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Wuhan, China
| | - Lixue Huang
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Pulmonary and Critical Care Medicine, Capital Medical University, Beijing, China
| | - Yeming Wang
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xia Li
- Department of COVID-19 Re-examination Clinic, Jin Yin-tan Hospital, Wuhan, China; Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Wuhan, China
| | - Lili Ren
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Key Laboratory of Respiratory Disease Pathogenomics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoying Gu
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China; Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liang Kang
- Medical Department, Jin Yin-tan Hospital, Wuhan, China; Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Wuhan, China
| | - Li Guo
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Key Laboratory of Respiratory Disease Pathogenomics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Min Liu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Xing Zhou
- Department of COVID-19 Re-examination Clinic, Jin Yin-tan Hospital, Wuhan, China; Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Wuhan, China
| | - Jianfeng Luo
- Department of COVID-19 Re-examination Clinic, Jin Yin-tan Hospital, Wuhan, China; Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Wuhan, China
| | - Zhenghui Huang
- Department of COVID-19 Re-examination Clinic, Jin Yin-tan Hospital, Wuhan, China; Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Wuhan, China
| | - Shengjin Tu
- Department of COVID-19 Re-examination Clinic, Jin Yin-tan Hospital, Wuhan, China; Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Wuhan, China
| | - Yue Zhao
- Department of COVID-19 Re-examination Clinic, Jin Yin-tan Hospital, Wuhan, China
| | - Li Chen
- Department of COVID-19 Re-examination Clinic, Jin Yin-tan Hospital, Wuhan, China
| | - Decui Xu
- Department of COVID-19 Re-examination Clinic, Jin Yin-tan Hospital, Wuhan, China
| | - Yanping Li
- Department of COVID-19 Re-examination Clinic, Jin Yin-tan Hospital, Wuhan, China
| | - Caihong Li
- Department of COVID-19 Re-examination Clinic, Jin Yin-tan Hospital, Wuhan, China
| | - Lu Peng
- Department of COVID-19 Re-examination Clinic, Jin Yin-tan Hospital, Wuhan, China
| | - Yong Li
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wuxiang Xie
- Peking University Clinical Research Institute, Beijing, China
| | - Dan Cui
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Harbin Medical University, Harbin, China
| | - Lianhan Shang
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Beijing University of Chinese Medicine, Beijing, China
| | - Guohui Fan
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China; Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiuyang Xu
- Tsinghua University School of Medicine, Beijing, China
| | - Geng Wang
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Key Laboratory of Respiratory Disease Pathogenomics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Wang
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Key Laboratory of Respiratory Disease Pathogenomics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingchuan Zhong
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Key Laboratory of Respiratory Disease Pathogenomics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chen Wang
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Tsinghua University-Peking University Joint Center for Life Sciences, Beijing, China
| | - Jianwei Wang
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Key Laboratory of Respiratory Disease Pathogenomics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dingyu Zhang
- Medical Department, Jin Yin-tan Hospital, Wuhan, China; Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Wuhan, China
| | - Bin Cao
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Pulmonary and Critical Care Medicine, Capital Medical University, Beijing, China; Tsinghua University-Peking University Joint Center for Life Sciences, Beijing, China.
| |
Collapse
|
81
|
Yu D, Han HJ, Yu J, Kim J, Lee GH, Yang JH, Song BM, Tark D, Choi BS, Kang SM, Heo WD. Pseudoknot-targeting Cas13b combats SARS-CoV-2 infection by suppressing viral replication. Mol Ther 2023; 31:1675-1687. [PMID: 36945774 PMCID: PMC10028249 DOI: 10.1016/j.ymthe.2023.03.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 02/25/2023] [Accepted: 03/16/2023] [Indexed: 03/23/2023] Open
Abstract
CRISPR-Cas13-mediated viral genome targeting is a novel strategy for defending against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants. Here, we generated mRNA-encoded Cas13b targeting the open reading frame 1b (ORF1b) region to effectively degrade the RNA-dependent RNA polymerase gene. Of the 12 designed CRISPR RNAs (crRNAs), those targeting the pseudoknot site upstream of ORF1b were found to be the most effective in suppressing SARS-CoV-2 propagation. Pseudoknot-targeting Cas13b reduced expression of the spike protein and attenuated viral replication by 99%. It also inhibited the replication of multiple SARS-CoV-2 variants, exhibiting broad potency. We validated the therapeutic efficacy of this system in SARS-CoV-2-infected hACE2 transgenic mice, demonstrating that crRNA treatment significantly reduced viral titers. Our findings suggest that the pseudoknot region is a strategic site for targeted genomic degradation of SARS-CoV-2. Hence, pseudoknot-targeting Cas13b could be a breakthrough therapy for overcoming infections by SARS-CoV-2 or other RNA viruses.
Collapse
Affiliation(s)
- Daseuli Yu
- Life Science Research Institute, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Hee-Jeong Han
- Laboratory for Infectious Disease Prevention, Korea Zoonosis Research Institute, Jeonbuk National University, Iksan 54531, Republic of Korea
| | - Jeonghye Yu
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jihye Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Gun-Hee Lee
- Laboratory for Infectious Disease Prevention, Korea Zoonosis Research Institute, Jeonbuk National University, Iksan 54531, Republic of Korea
| | - Ju-Hee Yang
- Laboratory for Infectious Disease Prevention, Korea Zoonosis Research Institute, Jeonbuk National University, Iksan 54531, Republic of Korea
| | - Byeong-Min Song
- Laboratory for Infectious Disease Prevention, Korea Zoonosis Research Institute, Jeonbuk National University, Iksan 54531, Republic of Korea
| | - Dongseob Tark
- Laboratory for Infectious Disease Prevention, Korea Zoonosis Research Institute, Jeonbuk National University, Iksan 54531, Republic of Korea
| | - Byeong-Sun Choi
- Honam Regional Center for Disease Control and Prevention, RCDC, Korea Disease Control and Prevention Agency, Gwangju 61947, Republic of Korea
| | - Sang-Min Kang
- Laboratory for Infectious Disease Prevention, Korea Zoonosis Research Institute, Jeonbuk National University, Iksan 54531, Republic of Korea.
| | - Won Do Heo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea.
| |
Collapse
|
82
|
van Dam KPJ, Volkers AG, Wieske L, Stalman EW, Kummer LYL, van Kempen ZLE, Killestein J, Tas SW, Boekel L, Wolbink GJ, van der Kooi AJ, Raaphorst J, Takkenberg RB, D'Haens GRAM, Spuls PI, Bekkenk MW, Musters AH, Post NF, Bosma AL, Hilhorst ML, Vegting Y, Bemelman FJ, Voskuyl AE, Broens B, Sanchez AP, van Els CACM, de Wit J, Rutgers A, de Leeuw K, Horváth B, Verschuuren JJGM, Ruiter AM, van Ouwerkerk L, van der Woude D, Allaart RCF, Teng YKO, van Paassen P, Busch MH, Jallah PBP, Brusse E, van Doorn PA, Baars AE, Hijnen DJ, Schreurs CRG, van der Pol WL, Goedee HS, Steenhuis M, Keijzer S, Keijser JBD, Cristianawati O, Ten Brinke A, Verstegen NJM, van Ham SM, Rispens T, Kuijpers TW, Löwenberg M, Eftimov F. Primary SARS-CoV-2 infection in patients with immune-mediated inflammatory diseases: long-term humoral immune responses and effects on disease activity. BMC Infect Dis 2023; 23:332. [PMID: 37198536 DOI: 10.1186/s12879-023-08298-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 04/29/2023] [Indexed: 05/19/2023] Open
Abstract
BACKGROUND Patients with immune-mediated inflammatory diseases (IMIDs) on immunosuppressants (ISPs) may have impaired long-term humoral immune responses and increased disease activity after SARS-CoV-2 infection. We aimed to investigate long-term humoral immune responses against SARS-CoV-2 and increased disease activity after a primary SARS-CoV-2 infection in unvaccinated IMID patients on ISPs. METHODS IMID patients on active treatment with ISPs and controls (i.e. IMID patients not on ISP and healthy controls) with a confirmed SARS-CoV-2 infection before first vaccination were included from an ongoing prospective cohort study (T2B! study). Clinical data on infections and increased disease activity were registered using electronic surveys and health records. A serum sample was collected before first vaccination to measure SARS-CoV-2 anti-receptor-binding domain (RBD) antibodies. RESULTS In total, 193 IMID patients on ISP and 113 controls were included. Serum samples from 185 participants were available, with a median time of 173 days between infection and sample collection. The rate of seropositive IMID patients on ISPs was 78% compared to 100% in controls (p < 0.001). Seropositivity rates were lowest in patients on anti-CD20 (40.0%) and anti-tumor necrosis factor (TNF) agents (60.5%), as compared to other ISPs (p < 0.001 and p < 0.001, respectively). Increased disease activity after infection was reported by 68 of 260 patients (26.2%; 95% CI 21.2-31.8%), leading to ISP intensification in 6 out of these 68 patients (8.8%). CONCLUSION IMID patients using ISPs showed reduced long-term humoral immune responses after primary SARS-CoV-2 infection, which was mainly attributed to treatment with anti-CD20 and anti-TNF agents. Increased disease activity after SARS-CoV-2 infection was reported commonly, but was mostly mild. TRIAL REGISTRATION NL74974.018.20, Trial ID: NL8900. Registered on 9 September 2020.
Collapse
Affiliation(s)
- Koos P J van Dam
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Adriaan G Volkers
- Department of Gastroenterology and Hepatology, Location Academic Medical Center, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Luuk Wieske
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
- Department of Clinical Neurophysiology, St Antonius Hospital, Nieuwegein, The Netherlands
| | - Eileen W Stalman
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Laura Y L Kummer
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, the Netherlands
| | - Zoé L E van Kempen
- Department of Neurology, Amsterdam UMC, VU University Medical Center, Amsterdam, the Netherlands
| | - Joep Killestein
- Department of Neurology, Amsterdam UMC, VU University Medical Center, Amsterdam, the Netherlands
| | - Sander W Tas
- Amsterdam Rheumatology and Immunology Center, Amsterdam UMC, Department of Rheumatology and Clinical Immunology, University of Amsterdam, Amsterdam, the Netherlands
| | - Laura Boekel
- Amsterdam Rheumatology and Immunology Center, Location Reade, Department of Rheumatology, Amsterdam, the Netherlands
| | - Gerrit J Wolbink
- Amsterdam Rheumatology and Immunology Center, Location Reade, Department of Rheumatology, Amsterdam, the Netherlands
| | - Anneke J van der Kooi
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Joost Raaphorst
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - R Bart Takkenberg
- Department of Gastroenterology and Hepatology, Location Academic Medical Center, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Geert R A M D'Haens
- Department of Gastroenterology and Hepatology, Location Academic Medical Center, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Phyllis I Spuls
- Department of Dermatology, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Marcel W Bekkenk
- Department of Dermatology, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Annelie H Musters
- Department of Dermatology, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Nicoline F Post
- Department of Dermatology, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Angela L Bosma
- Department of Dermatology, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Marc L Hilhorst
- Department of Internal Medicine, Section of Nephrology, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Yosta Vegting
- Department of Internal Medicine, Section of Nephrology, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Frederike J Bemelman
- Department of Internal Medicine, Section of Nephrology, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Alexandre E Voskuyl
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and Immunology Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Bo Broens
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and Immunology Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Agner Parra Sanchez
- Amsterdam Rheumatology and Immunology Center, Amsterdam UMC, Department of Rheumatology and Clinical Immunology, University of Amsterdam, Amsterdam, the Netherlands
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and Immunology Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Cécile A C M van Els
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
- Faculty of Veterinary Medicine, Utrecht University Utrecht, Utrecht, The Netherlands
| | - Jelle de Wit
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Abraham Rutgers
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, Groningen, The Netherlands
| | - Karina de Leeuw
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, Groningen, The Netherlands
| | - Barbara Horváth
- Department of Dermatology, Center for Blistering Diseases, University Medical Center Groningen, University Groningen, Groningen, The Netherlands
| | | | - Annabel M Ruiter
- Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands
| | - Lotte van Ouwerkerk
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Diane van der Woude
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Renée C F Allaart
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Y K Onno Teng
- Centre of Expertise for Lupus-, Vasculitis- and Complement-Mediated Systemic Diseases, Department of Internal Medicine - Nephrology section, Leiden University Medical Centre, Leiden, The Netherlands
| | - Pieter van Paassen
- Department of Nephrology and Clinical Immunology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Matthias H Busch
- Department of Nephrology and Clinical Immunology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Papay B P Jallah
- Department of Nephrology and Clinical Immunology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Esther Brusse
- Department of Neurology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Pieter A van Doorn
- Department of Neurology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Adája E Baars
- Department of Neurology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Dirk Jan Hijnen
- Department of Dermatology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Corine R G Schreurs
- Department of Dermatology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - W Ludo van der Pol
- Department of Neurology and Neurosurgery, Brain Center UMC Utrecht, Utrecht, the Netherlands
| | - H Stephan Goedee
- Department of Neurology and Neurosurgery, Brain Center UMC Utrecht, Utrecht, the Netherlands
| | - Maurice Steenhuis
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, the Netherlands
| | - Sofie Keijzer
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, the Netherlands
| | - Jim B D Keijser
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, the Netherlands
| | - Olvi Cristianawati
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, the Netherlands
| | - Anja Ten Brinke
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, the Netherlands
| | - Niels J M Verstegen
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, the Netherlands
| | - S Marieke van Ham
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, the Netherlands
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, the Netherlands
| | - Taco W Kuijpers
- Department of Pediatric Immunology, Rheumatology and Infectious Disease, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Mark Löwenberg
- Department of Gastroenterology and Hepatology, Location Academic Medical Center, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Filip Eftimov
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands.
| |
Collapse
|
83
|
Ng HM, Lei CL, Fu S, Li E, Leong SI, Nip CI, Choi NM, Lai KS, Tang XJ, Lei CL, Xu RH. Heterologous vaccination with inactivated vaccine and mRNA vaccine augments antibodies against both spike and nucleocapsid proteins of SARS-CoV-2: a local study in Macao. Front Immunol 2023; 14:1131985. [PMID: 37251391 PMCID: PMC10213252 DOI: 10.3389/fimmu.2023.1131985] [Citation(s) in RCA: 2] [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: 12/26/2022] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
The mRNA vaccines (RVs) can reduce the severity and mortality of severe acute respiratory syndrome coronavirus (SARS-CoV-2). However, almost only the inactivated vaccines (IVs) but no RVs had been used in mainland China until most recently, and the relaxing of its anti-pandemic strategies in December 2022 increased concerns about new outbreaks. In comparison, many of the citizens in Macao Special Administrative Region of China received three doses of IV (3IV) or RV (3RV), or 2 doses of IV plus one booster of RV (2IV+1RV). By the end of 2022, we recruited 147 participants with various vaccinations in Macao and detected antibodies (Abs) against the spike (S) protein and nucleocapsid (N) protein of the virus as well as neutralizing antibodies (NAb) in their serum. We observed that the level of anti-S Ab or NAb was similarly high with both 3RV and 2IV+1RV but lower with 3IV. In contrast, the level of anti-N Ab was the highest with 3IV like that in convalescents, intermediate with 2IV+1RV, and the lowest with 3RV. Whereas no significant differences in the basal levels of cytokines related to T-cell activation were observed among the various vaccination groups before and after the boosters. No vaccinees reported severe adverse events. Since Macao took one of the most stringent non-pharmaceutical interventions in the world, this study possesses much higher confidence in the vaccination results than many other studies from highly infected regions. Our findings suggest that the heterologous vaccination 2IV+1RV outperforms the homologous vaccinations 3IV and 3RV as it induces not only anti-S Ab (to the level as with 3RV) but also anti-N antibodies (via the IV). It combines the advantages of both RV (to block the viral entry) and IV (to also intervene the subsequent pathological processes such as intracellular viral replication and interference with the signal transduction and hence the biological functions of host cells).
Collapse
Affiliation(s)
- Hoi Man Ng
- Laboratory Department, Kiang Wu Hospital, Macao, Macao SAR, China
| | - Chon Lok Lei
- Faculty of Health Sciences, University of Macau, Macao, Macao SAR, China
| | - Siyi Fu
- Faculty of Health Sciences, University of Macau, Macao, Macao SAR, China
| | - Enqin Li
- Faculty of Health Sciences, University of Macau, Macao, Macao SAR, China
| | - Sek In Leong
- Laboratory Department, Kiang Wu Hospital, Macao, Macao SAR, China
| | - Chu Iong Nip
- Laboratory Department, Kiang Wu Hospital, Macao, Macao SAR, China
| | - Nga Man Choi
- Laboratory Department, Kiang Wu Hospital, Macao, Macao SAR, China
| | - Kai Seng Lai
- Laboratory Department, Kiang Wu Hospital, Macao, Macao SAR, China
| | - Xi Jun Tang
- Laboratory Department, Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, Zhuhai, China
| | - Chon Leng Lei
- Laboratory Department, Kiang Wu Hospital, Macao, Macao SAR, China
| | - Ren-He Xu
- Faculty of Health Sciences, University of Macau, Macao, Macao SAR, China
| |
Collapse
|
84
|
Shen J, Fan J, Zhao Y, Jiang D, Niu Z, Zhang Z, Cao G. Innate and adaptive immunity to SARS-CoV-2 and predisposing factors. Front Immunol 2023; 14:1159326. [PMID: 37228604 PMCID: PMC10203583 DOI: 10.3389/fimmu.2023.1159326] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 04/27/2023] [Indexed: 05/27/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus (SARS-CoV-2), has affected all countries worldwide. Although some symptoms are relatively mild, others are still associated with severe and even fatal clinical outcomes. Innate and adaptive immunity are important for the control of SARS-CoV-2 infections, whereas a comprehensive characterization of the innate and adaptive immune response to COVID-19 is still lacking and the mechanisms underlying immune pathogenesis and host predisposing factors are still a matter of scientific debate. Here, the specific functions and kinetics of innate and adaptive immunity involved in SARS-CoV-2 recognition and resultant pathogenesis are discussed, as well as their immune memory for vaccinations, viral-mediated immune evasion, and the current and future immunotherapeutic agents. We also highlight host factors that contribute to infection, which may deepen the understanding of viral pathogenesis and help identify targeted therapies that attenuate severe disease and infection.
Collapse
Affiliation(s)
- Jiaying Shen
- Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Junyan Fan
- Department of Epidemiology, Shanghai Key Laboratory of Medical Bioprotection, Key Laboratory of Biological Defense, Ministry of Education, Second Military Medical University, Shanghai, China
| | - Yue Zhao
- Department of Epidemiology, Shanghai Key Laboratory of Medical Bioprotection, Key Laboratory of Biological Defense, Ministry of Education, Second Military Medical University, Shanghai, China
| | - Doming Jiang
- Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Zheyun Niu
- Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Zihan Zhang
- Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Guangwen Cao
- Tongji University School of Medicine, Tongji University, Shanghai, China
- Department of Epidemiology, Shanghai Key Laboratory of Medical Bioprotection, Key Laboratory of Biological Defense, Ministry of Education, Second Military Medical University, Shanghai, China
| |
Collapse
|
85
|
Cai C, Li Y, Hu T, Liang R, Wang K, Guo C, Li Y, Zhang M, Kang M. The Associated Factors of SARS-CoV-2 Reinfection by Omicron Variant - Guangdong Province, China, December 2022 to January 2023. China CDC Wkly 2023; 5:391-396. [PMID: 37197174 PMCID: PMC10184471 DOI: 10.46234/ccdcw2023.075] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/11/2023] [Indexed: 05/19/2023] Open
Abstract
What is already known about this topic? Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) reinfection by variants is being reported commonly and has caused waves of epidemic in many countries. Because of dynamic zero policy, the SARS-CoV-2 reinfection was less reported in China. What is added by this report? SARS-CoV-2 reinfections were observed in Guangdong Province between December 2022 and January 2023. This study estimated that the reinfection incidence was 50.0% for the original strain primary infections, 35.2% for the Alpha or Delta variants, and 18.4% for the Omicron variant; The reinfection incidence within 3-6 months after primary infection by Omicron variant was 4.0%. Besides, 96.2% reinfection cases were symptomatic while only 7.7% sought medical attention. What are the implications for public health practice? These findings suggest a reduced likelihood of an Omicron-driven epidemic resurgence in the short term but emphasize the importance of maintaining vigilant surveillance of emerging SARS-CoV-2 variants and conducting population-based antibody level surveys to inform response preparedness.
Collapse
Affiliation(s)
- Chunsheng Cai
- Guangdong Field Epidemiology Training Program, Guangzhou City, Guangdong Province, China
- Zhongshan Center for Disease Control and Prevention, Zhongshan City, Guangdong Province, China
| | - Yihong Li
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou City, Guangdong Province, China
| | - Ting Hu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou City, Guangdong Province, China
| | - Rongwei Liang
- Guangdong Field Epidemiology Training Program, Guangzhou City, Guangdong Province, China
- Huaiji County Center for Disease Control and Prevention, Zhaoqing City, Guangdong Province, China
| | - Kaibin Wang
- Guangdong Field Epidemiology Training Program, Guangzhou City, Guangdong Province, China
- Tianhe District Center for Disease Control and Prevention, Guangzhou City, Guangdong Province, China
| | - Congrui Guo
- Guangdong Field Epidemiology Training Program, Guangzhou City, Guangdong Province, China
- Futian District Center for Disease Control and Prevention, Shenzhen City, Guangdong Province, China
| | - Yan Li
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou City, Guangdong Province, China
| | - Meng Zhang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou City, Guangdong Province, China
| | - Min Kang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou City, Guangdong Province, China
- Min Kang,
| |
Collapse
|
86
|
Ören MM, Canbaz S, Meşe S, Ağaçfidan A, Demir ÖS, Karaca E, Doğruyol AR, Otçu GH, Tükek T, Özgülnar N. Impact of Health Workers' Choice of COVID-19 Vaccine Booster on Immunization Levels in Istanbul, Turkey. Vaccines (Basel) 2023; 11:vaccines11050935. [PMID: 37243039 DOI: 10.3390/vaccines11050935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND There are limited data regarding short- and medium-term IgG antibody levels after the CoronaVac and BNT162b2 vaccines. This study aimed to investigate the antibody responses of health workers who initially received two doses of CoronaVac one month apart followed by a booster dose of either CoronaVac or BNT162b2, as well as determine whether either vaccine provided superior results. METHODS This research represents the second phase of a mixed-methods vaccine cohort study and was conducted between July 2021 and February 2022. The participants (n = 117) were interviewed in person and blood samples were collected before and at 1 and 6 months after the booster vaccination. RESULTS BNT162b2 was found to have greater immunogenic potential than CoronaVac (p < 0.001). Health workers without chronic disease exhibited statistically significant increases in antibody levels after both vaccines (p < 0.001), whereas only BNT162b2 caused a significant increase in antibody levels in participants with chronic disease (p < 0.001). Samples obtained before and at 1 and 6 months after the booster vaccination revealed no age- or sex-based differences in IgG-inducing potential for either vaccine (p > 0.05). Antibody levels were comparable in both vaccine groups before the booster regardless of COVID-19 history (p > 0.05); however, antibody levels were significantly higher after the BNT162b2 booster at 1 month (<0.001) and at 6 months, except among participants who had a positive history of COVID-19 infection (p < 0.001). CONCLUSIONS Our results suggest that even a single booster dose of BNT162b2 after initial vaccination with CoronaVac provides a protective advantage against COVID-19, especially for risk groups such as health workers and those with chronic diseases.
Collapse
Affiliation(s)
- Meryem Merve Ören
- Department of Public Health, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34116, Turkey
| | - Sevgi Canbaz
- Department of Public Health, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34116, Turkey
| | - Sevim Meşe
- Department of Medical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34116, Turkey
| | - Ali Ağaçfidan
- Department of Medical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34116, Turkey
| | - Ömer Serdil Demir
- Department of Public Health, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34116, Turkey
| | - Esra Karaca
- Department of Public Health, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34116, Turkey
| | - Ayşe Rumeysa Doğruyol
- Department of Public Health, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34116, Turkey
| | - Gökçe Hazar Otçu
- Department of Public Health, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34116, Turkey
| | - Tufan Tükek
- Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34116, Turkey
| | - Nuray Özgülnar
- Department of Public Health, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34116, Turkey
| |
Collapse
|
87
|
Peng M, Hu M, Peng X, Gong Y, Qian K, Li J, Zhao J, Li X, Huang J, Zhang M, Chai L, Chen L, Zhang D, Peng L. What contributes to the re-positive nucleic acid test results for the omicron variant of SARS-CoV-2 in the shelter cabin hospital in Shanghai, China? Heliyon 2023; 9:e15679. [PMID: 37124338 PMCID: PMC10123020 DOI: 10.1016/j.heliyon.2023.e15679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 04/18/2023] [Accepted: 04/18/2023] [Indexed: 05/02/2023] Open
Abstract
Background Despite the increasing reports of re-positive SARS-CoV-2 cases after recovery and discharge from hospitals, our knowledge remains very limited regarding the contributing factors of re-positivity and its roles in the transmission and epidemiology of the Omicron variant. Methods In this retrospective study, re-positivity is defined as the positive nucleic acid result (Ct < 35) following two consecutive negative results during hospitalization. A total of 751 patients from Shanghai Shelter Cabin Hospital were enrolled and divided with a ratio of about 1:2 into the re-positivity group and the non-re-positivity group. Patients required three consecutive negative results daily as the de-isolation criterion. The follow-up time of discharged patients lasted five weeks. Univariate regression analysis was used to compare variables between the re-positivity and non-re-positivity groups, and the single re-positivity and multiple re-positivity groups, with P < 0.05 defined as the statistical significance of differences. Subsequently, variables with P < 0.2 were subjected to multivariate logistic regression analysis to investigate the odds ratio (OR) of re-positivity and the influencing factors of re-positivity of the Omicron variant. Results The re-positivity group had a higher proportion of males (68.1% vs 58.1%, p = 0.000), a higher education level (31.9% vs 12.7%, p = 0.007), a longer hospitalization duration (13 days vs 8 days, p = 0.000), and a higher Convidecia vaccination rate (6.0% vs 2.4%, p = 0.011). Further multivariable analysis showed male (OR = 2.168, p = 0.000), Convidecia vaccination (OR = 2.634, p = 0.014), hospitalization duration (OR = 2.146, p = 0.000) and education level (OR = 1.595, p = 0.007) were associated with re-positivity. The average rate of re-positivity was 25% during hospitalization and decreased to 0.4% among discharged patients. Re-positivity was more common in the period with a larger number of hospitalized patients and in larger wards with a larger number of patients. Conclusion A large number of hospitalized patients, large-sized wards, and gender are significant contributing factors to re-positivity. Division of the shelter cabin hospital into small independent wards and requirement of three consecutive results daily as the de-isolation criterion might be more beneficial to the control and prevention of the spread of the Omicron variant.
Collapse
Affiliation(s)
- Mei Peng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Maozi Hu
- Gastroenterology Department, Jiulongpo District Hospital of Chongqing, Chongqing, China
| | - Xiaolu Peng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuan Gong
- Critical Care Medicine Department, Jiulongpo District Hospital of Chongqing, Chongqing, China
| | - Keli Qian
- Department of Nosocomial Infection Control, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Junnan Li
- Department of Obstetrics and Fetal Medicine Unit, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jinqiu Zhao
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiang Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Huang
- Department of Respiratory Medicine, Chongqing General Hospital, Chongqing, China
| | - Meng Zhang
- Department of Critical Care Medicine, Chongqing General Hospital, Chongqing, China
| | - Lili Chai
- Infectious Department, The Fifth Rehabilitation Hospital of Shanghai, Shanghai, China
| | - Li Chen
- Medical Service Division, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dan Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Peng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
88
|
Shamseldin MM, Kenney A, Zani A, Evans JP, Zeng C, Read KA, Hall JM, Chaiwatpongsakorn S, Mahesh KC, Lu M, Eltobgy M, Denz P, Deora R, Li J, Peeples ME, Oestreich KJ, Liu SL, Corps KN, Yount JS, Dubey P. Prime-Pull Immunization of Mice with a BcfA-Adjuvanted Vaccine Elicits Sustained Mucosal Immunity That Prevents SARS-CoV-2 Infection and Pathology. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1257-1271. [PMID: 36881867 PMCID: PMC10121870 DOI: 10.4049/jimmunol.2200297] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 02/15/2023] [Indexed: 03/09/2023]
Abstract
Vaccines against SARS-CoV-2 that induce mucosal immunity capable of preventing infection and disease remain urgently needed. In this study, we demonstrate the efficacy of Bordetella colonization factor A (BcfA), a novel bacteria-derived protein adjuvant, in SARS-CoV-2 spike-based prime-pull immunizations. We show that i.m. priming of mice with an aluminum hydroxide- and BcfA-adjuvanted spike subunit vaccine, followed by a BcfA-adjuvanted mucosal booster, generated Th17-polarized CD4+ tissue-resident memory T cells and neutralizing Abs. Immunization with this heterologous vaccine prevented weight loss following challenge with mouse-adapted SARS-CoV-2 (MA10) and reduced viral replication in the respiratory tract. Histopathology showed a strong leukocyte and polymorphonuclear cell infiltrate without epithelial damage in mice immunized with BcfA-containing vaccines. Importantly, neutralizing Abs and tissue-resident memory T cells were maintained until 3 mo postbooster. Viral load in the nose of mice challenged with the MA10 virus at this time point was significantly reduced compared with naive challenged mice and mice immunized with an aluminum hydroxide-adjuvanted vaccine. We show that vaccines adjuvanted with alum and BcfA, delivered through a heterologous prime-pull regimen, provide sustained protection against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Mohamed M Shamseldin
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
- Department of Microbiology, The Ohio State University, Columbus, OH
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University-Ain Helwan, Helwan, Egypt
| | - Adam Kenney
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| | - Ashley Zani
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| | - John P Evans
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH
- Center for Retrovirus Research, The Ohio State University, Columbus, OH
| | - Cong Zeng
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH
- Center for Retrovirus Research, The Ohio State University, Columbus, OH
| | - Kaitlin A Read
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| | - Jesse M Hall
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| | - Supranee Chaiwatpongsakorn
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH
| | - K C Mahesh
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH
| | - Mijia Lu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH
| | - Mostafa Eltobgy
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| | - Parker Denz
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| | - Rajendar Deora
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
- Department of Microbiology, The Ohio State University, Columbus, OH
| | - Jianrong Li
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH
| | - Mark E Peeples
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH
- Department of Pediatrics, The Ohio State University, Columbus, OH
| | - Kenneth J Oestreich
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| | - Shan-Lu Liu
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
- Department of Microbiology, The Ohio State University, Columbus, OH
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH
- Center for Retrovirus Research, The Ohio State University, Columbus, OH
| | - Kara N Corps
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH
| | - Jacob S Yount
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| | - Purnima Dubey
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| |
Collapse
|
89
|
Miyazato Y, Tsuzuki S, Matsunaga A, Morioka S, Terada M, Saito S, Iwamoto N, Kutsuna S, Ishizaka Y, Ohmagari N. Association between SARS-CoV-2 anti-spike antibody titers and the development of post-COVID conditions: A retrospective observational study. Glob Health Med 2023; 5:106-111. [PMID: 37128226 PMCID: PMC10130543 DOI: 10.35772/ghm.2022.01070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 04/04/2023] [Accepted: 04/07/2023] [Indexed: 05/03/2023]
Abstract
The symptoms that persist after an acute coronavirus disease 2019 (COVID-19) are referred to as post- COVID conditions. Although the cause of post-COVID conditions remains unclear, the host immune response to SARS-CoV-2 may be involved. Hence, we aimed to investigate the effect of serum antibody titers against SARS-CoV-2 on the development of post-COVID conditions. We conducted a retrospective observational study of COVID-19-recovered individuals who attended the clinic at the National Center for Global Health and Medicine between January 2020 and April 2021. Serum SARS-CoV-2 anti-spike antibody titers were measured and a questionnaire survey was used to collect information on the presence of post-COVID conditions and demographic characteristics of the participants. Participants were then divided into two groups: high peak antibody titer group [≥ 0.759 OD450 value], and low peak antibody titer group [< 0.759 OD450 value] and compared their frequency of post-COVID conditions. Of 526 individuals attending the clinic, 457 (86.9%) responded to the questionnaire. We analyzed the data of 227 (49.7%) participants with measurements of serum antibody titers during the peak period. The incidence of depressed mood was significantly higher in the group with higher antibody titers (odds ratio: 2.34, 95% CI: 1.17-4.67, p = 0.016). There was no significant difference in the frequency of the remaining symptoms between the two groups. Among post-COVID conditions, the depressed mood was more frequent in the group with high serum antibody titers which suggests a difference in pathogenesis between depressive mood and other post-COVID conditions that requires further investigation.
Collapse
Affiliation(s)
- Yusuke Miyazato
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shinya Tsuzuki
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
- AMR Clinical Reference Center, National Center for Global Health and Medicine Hospital, Tokyo, Japan
- Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Akihiro Matsunaga
- Department of Intractable Diseases, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shinichiro Morioka
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
- AMR Clinical Reference Center, National Center for Global Health and Medicine Hospital, Tokyo, Japan
- Emerging and Reemerging Infectious Diseases, Graduate School of Medicine, Tohoku University, Miyagi, Japan
- Address correspondence to:Shinichiro Morioka, Disease Control and Prevention Center, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan. E-mail:
| | - Mari Terada
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
- Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Sho Saito
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Noriko Iwamoto
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Satoshi Kutsuna
- Department of Infection Control, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yukihito Ishizaka
- Department of Intractable Diseases, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Norio Ohmagari
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
- AMR Clinical Reference Center, National Center for Global Health and Medicine Hospital, Tokyo, Japan
| |
Collapse
|
90
|
Kenny G, Townsend L, Savinelli S, Mallon PWG. Long COVID: Clinical characteristics, proposed pathogenesis and potential therapeutic targets. Front Mol Biosci 2023; 10:1157651. [PMID: 37179568 PMCID: PMC10171433 DOI: 10.3389/fmolb.2023.1157651] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/04/2023] [Indexed: 05/15/2023] Open
Abstract
The emergence of persistent ill-health in the aftermath of SARS-CoV-2 infection has presented significant challenges to patients, healthcare workers and researchers. Termed long COVID, or post-acute sequelae of COVID-19 (PASC), the symptoms of this condition are highly variable and span multiple body systems. The underlying pathophysiology remains poorly understood, with no therapeutic agents proven to be effective. This narrative review describes predominant clinical features and phenotypes of long COVID alongside the data supporting potential pathogenesis of these phenotypes including ongoing immune dysregulation, viral persistence, endotheliopathy, gastrointestinal microbiome disturbance, autoimmunity, and dysautonomia. Finally, we describe current potential therapies under investigation, as well as future potential therapeutic options based on the proposed pathogenesis research.
Collapse
Affiliation(s)
- Grace Kenny
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
- Department of Infectious Diseases, St Vincent’s University Hospital, Dublin, Ireland
| | - Liam Townsend
- Department of Infectious Diseases, St Vincent’s University Hospital, Dublin, Ireland
| | - Stefano Savinelli
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
- Department of Infectious Diseases, St Vincent’s University Hospital, Dublin, Ireland
| | - Patrick W. G. Mallon
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
- Department of Infectious Diseases, St Vincent’s University Hospital, Dublin, Ireland
| |
Collapse
|
91
|
Sunagar R, Singh A, Kumar S. SARS-CoV-2: Immunity, Challenges with Current Vaccines, and a Novel Perspective on Mucosal Vaccines. Vaccines (Basel) 2023; 11:vaccines11040849. [PMID: 37112761 PMCID: PMC10143972 DOI: 10.3390/vaccines11040849] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
The global rollout of COVID-19 vaccines has played a critical role in reducing pandemic spread, disease severity, hospitalizations, and deaths. However, the first-generation vaccines failed to block severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and transmission, partially due to the limited induction of mucosal immunity, leading to the continuous emergence of variants of concern (VOC) and breakthrough infections. To meet the challenges from VOC, limited durability, and lack of mucosal immune response of first-generation vaccines, novel approaches are being investigated. Herein, we have discussed the current knowledge pertaining to natural and vaccine-induced immunity, and the role of the mucosal immune response in controlling SARS-CoV2 infection. We have also presented the current status of the novel approaches aimed at eliciting both mucosal and systemic immunity. Finally, we have presented a novel adjuvant-free approach to elicit effective mucosal immunity against SARS-CoV-2, which lacks the safety concerns associated with live-attenuated vaccine platforms.
Collapse
Affiliation(s)
| | - Amit Singh
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA
| | - Sudeep Kumar
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA
| |
Collapse
|
92
|
Wang J, Huang L, Guo N, Yao YP, Zhang C, Xu R, Jiao YM, Li YQ, Song YR, Wang FS, Fan X. Dynamics of SARS-CoV-2 Antibody Responses up to 9 Months Post-Vaccination in Individuals with Previous SARS-CoV-2 Infection Receiving Inactivated Vaccines. Viruses 2023; 15:v15040917. [PMID: 37112897 PMCID: PMC10145073 DOI: 10.3390/v15040917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/20/2023] [Accepted: 03/24/2023] [Indexed: 04/07/2023] Open
Abstract
Humoral immunity confers protection against COVID-19. The longevity of antibody responses after receiving an inactivated vaccine in individuals with previous SARS-CoV-2 infection is unclear. Plasma samples were collected from 58 individuals with previous SARS-CoV-2 infection and 25 healthy donors (HDs) who had been vaccinated with an inactivated vaccine. The neutralizing antibodies (NAbs) and S1 domain-specific antibodies against the SARS-CoV-2 wild-type and Omicron strains and nucleoside protein (NP)-specific antibodies were measured using a chemiluminescent immunoassay. Statistical analysis was performed using clinical variables and antibodies at different timepoints after SARS-CoV-2 vaccination. NAbs targeting the wild-type or Omicron strain were detected in individuals with previous SARS-CoV-2 infection at 12 months after infection (wild-type: 81%, geometric mean (GM): 20.3 AU/mL; Omicron: 44%, GM: 9.4 AU/mL), and vaccination provided further enhancement of these antibody levels (wild-type: 98%, GM: 53.3 AU/mL; Omicron: 75%, GM: 27.8 AU/mL, at 3 months after vaccination), which were significantly higher than those in HDs receiving a third dose of inactivated vaccine (wild-type: 85%, GM: 33.6 AU/mL; Omicron: 45%, GM: 11.5 AU/mL). The level of NAbs in individuals with previous infection plateaued 6 months after vaccination, but the NAb levels in HDs declined continuously. NAb levels in individuals with previous infection at 3 months post-vaccination were strongly correlated with those at 6 months post-vaccination, and weakly correlated with those before vaccination. NAb levels declined substantially in most individuals, and the rate of antibody decay was negatively correlated with the neutrophil-to-lymphocyte ratio in the blood at discharge. These results suggest that the inactivated vaccine induced robust and durable NAb responses in individuals with previous infection up to 9 months after vaccination.
Collapse
Affiliation(s)
- Jing Wang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China
| | - Lei Huang
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China
| | - Nan Guo
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China
- Chinese PLA Medical School, Beijing 100853, China
| | - Ya-Ping Yao
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
- Senior Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Chao Zhang
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China
| | - Ruonan Xu
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China
| | - Yan-Mei Jiao
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China
| | - Ya-Qun Li
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China
| | - Yao-Ru Song
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China
- Chinese PLA Medical School, Beijing 100853, China
| | - Fu-Sheng Wang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China
| | - Xing Fan
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China
| |
Collapse
|
93
|
Tangye SG. Impact of SARS-CoV-2 infection and COVID-19 on patients with inborn errors of immunity. J Allergy Clin Immunol 2023; 151:818-831. [PMID: 36522221 PMCID: PMC9746792 DOI: 10.1016/j.jaci.2022.11.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 12/15/2022]
Abstract
Since the arrival of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in December 2019, its characterization as a novel human pathogen, and the resulting coronavirus disease 2019 (COVID-19) pandemic, over 6.5 million people have died worldwide-a stark and sobering reminder of the fundamental and nonredundant roles of the innate and adaptive immune systems in host defense against emerging pathogens. Inborn errors of immunity (IEI) are caused by germline variants, typically in single genes. IEI are characterized by defects in development and/or function of cells involved in immunity and host defense, rendering individuals highly susceptible to severe, recurrent, and sometimes fatal infections, as well as immune dysregulatory conditions such as autoinflammation, autoimmunity, and allergy. The study of IEI has revealed key insights into the molecular and cellular requirements for immune-mediated protection against infectious diseases. Indeed, this has been exemplified by assessing the impact of SARS-CoV-2 infection in individuals with previously diagnosed IEI, as well as analyzing rare cases of severe COVID-19 in otherwise healthy individuals. This approach has defined fundamental aspects of mechanisms of disease pathogenesis, immunopathology in the context of infection with a novel pathogen, and therapeutic options to mitigate severe disease. This review summarizes these findings and illustrates how the study of these rare experiments of nature can inform key features of human immunology, which can then be leveraged to improve therapies for treating emerging and established infectious diseases.
Collapse
Affiliation(s)
- Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst, Darlinghurst, Australia; St Vincent's Clinical School, University of New South Wales Sydney, Randwick, Randwick, Australia; Clinical Immunogenomics Research Consortium of Australasia (CIRCA).
| |
Collapse
|
94
|
O’Connor MA, Hawman DW, Meade-White K, Leventhal S, Song W, Randall S, Archer J, Lewis TB, Brown B, Fredericks MN, Sprouse KR, Tunggal HC, Maughan M, Iwayama N, Ahrens C, Garrison W, Wangari S, Guerriero KA, Hanley P, Lovaglio J, Saturday G, Veesler D, Edlefsen PT, Khandhar AP, Feldmann H, Fuller DH, Erasmus JH. A replicon RNA vaccine can induce durable protective immunity from SARS-CoV-2 in nonhuman primates after neutralizing antibodies have waned. PLoS Pathog 2023; 19:e1011298. [PMID: 37075079 PMCID: PMC10150980 DOI: 10.1371/journal.ppat.1011298] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 05/01/2023] [Accepted: 04/11/2023] [Indexed: 04/20/2023] Open
Abstract
The global SARS-CoV-2 pandemic prompted rapid development of COVID-19 vaccines. Although several vaccines have received emergency approval through various public health agencies, the SARS-CoV-2 pandemic continues. Emergent variants of concern, waning immunity in the vaccinated, evidence that vaccines may not prevent transmission and inequity in vaccine distribution have driven continued development of vaccines against SARS-CoV-2 to address these public health needs. In this report, we evaluated a novel self-amplifying replicon RNA vaccine against SARS-CoV-2 in a pigtail macaque model of COVID-19 disease. We found that this vaccine elicited strong binding and neutralizing antibody responses against homologous virus. We also observed broad binding antibody against heterologous contemporary and ancestral strains, but neutralizing antibody responses were primarily targeted to the vaccine-homologous strain. While binding antibody responses were sustained, neutralizing antibody waned to undetectable levels in some animals after six months but were rapidly recalled and conferred protection from disease when the animals were challenged 7 months after vaccination as evident by reduced viral replication and pathology in the lower respiratory tract, reduced viral shedding in the nasal cavity and lower concentrations of pro-inflammatory cytokines in the lung. Cumulatively, our data demonstrate in pigtail macaques that a self-amplifying replicon RNA vaccine can elicit durable and protective immunity to SARS-CoV-2 infection. Furthermore, these data provide evidence that this vaccine can provide durable protective efficacy and reduce viral shedding even after neutralizing antibody responses have waned to undetectable levels.
Collapse
Affiliation(s)
- Megan A. O’Connor
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- Washington National Primate Research Center, University of Washington, Seattle, Washington, United States of America
| | - David W. Hawman
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana, United States of America
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana, United States of America
| | - Kimberly Meade-White
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana, United States of America
| | - Shanna Leventhal
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana, United States of America
| | - Wenjun Song
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Samantha Randall
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- HDT Bio, Seattle, Washington, United States of America
| | - Jacob Archer
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- HDT Bio, Seattle, Washington, United States of America
| | - Thomas B. Lewis
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- Washington National Primate Research Center, University of Washington, Seattle, Washington, United States of America
| | - Brieann Brown
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- Washington National Primate Research Center, University of Washington, Seattle, Washington, United States of America
| | - Megan N. Fredericks
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- Washington National Primate Research Center, University of Washington, Seattle, Washington, United States of America
| | - Kaitlin R. Sprouse
- Department of Biochemistry, University of Washington, United States of America
| | - Hillary C. Tunggal
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- Washington National Primate Research Center, University of Washington, Seattle, Washington, United States of America
| | - Mara Maughan
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- Washington National Primate Research Center, University of Washington, Seattle, Washington, United States of America
| | - Naoto Iwayama
- Washington National Primate Research Center, University of Washington, Seattle, Washington, United States of America
| | - Chul Ahrens
- Washington National Primate Research Center, University of Washington, Seattle, Washington, United States of America
| | - William Garrison
- Washington National Primate Research Center, University of Washington, Seattle, Washington, United States of America
| | - Solomon Wangari
- Washington National Primate Research Center, University of Washington, Seattle, Washington, United States of America
| | - Kathryn A. Guerriero
- Washington National Primate Research Center, University of Washington, Seattle, Washington, United States of America
| | - Patrick Hanley
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana, United States of America
| | - Jamie Lovaglio
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana, United States of America
| | - Greg Saturday
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana, United States of America
| | - David Veesler
- Department of Biochemistry, University of Washington, United States of America
| | - Paul T. Edlefsen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | | | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana, United States of America
| | - Deborah Heydenburg Fuller
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- Washington National Primate Research Center, University of Washington, Seattle, Washington, United States of America
| | - Jesse H. Erasmus
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- HDT Bio, Seattle, Washington, United States of America
| |
Collapse
|
95
|
Amanat F, Clark J, Carreño JM, Strohmeier S, Yellin T, Meade PS, Bhavsar D, Muramatsu H, Sun W, Coughlan L, Pardi N, Krammer F. Immunity to Seasonal Coronavirus Spike Proteins Does Not Protect from SARS-CoV-2 Challenge in a Mouse Model but Has No Detrimental Effect on Protection Mediated by COVID-19 mRNA Vaccination. J Virol 2023; 97:e0166422. [PMID: 36779758 PMCID: PMC10062180 DOI: 10.1128/jvi.01664-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 01/14/2023] [Indexed: 02/14/2023] Open
Abstract
Seasonal coronaviruses have been circulating widely in the human population for many years. With increasing age, humans are more likely to have been exposed to these viruses and to have developed immunity against them. It has been hypothesized that this immunity to seasonal coronaviruses may provide partial protection against infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and it has also been shown that coronavirus disease 2019 (COVID-19) vaccination induces a back-boosting effects against the spike proteins of seasonal betacoronaviruses. In this study, we tested if immunity to the seasonal coronavirus spikes from OC43, HKU1, 229E, or NL63 would confer protection against SARS-CoV-2 challenge in a mouse model, and whether pre-existing immunity against these spikes would weaken the protection afforded by mRNA COVID-19 vaccination. We found that mice vaccinated with the seasonal coronavirus spike proteins had no increased protection compared to the negative controls. While a negligible back-boosting effect against betacoronavirus spike proteins was observed after SARS-CoV-2 infection, there was no negative original antigenic sin-like effect on the immune response and protection induced by SARS-CoV-2 mRNA vaccination in animals with pre-existing immunity to seasonal coronavirus spike proteins. IMPORTANCE The impact that immunity against seasonal coronaviruses has on both susceptibility to SARS-CoV-2 infection as well as on COVID-19 vaccination is unclear. This study provides insights into both questions in a mouse model of SARS-CoV-2.
Collapse
Affiliation(s)
- Fatima Amanat
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jordan Clark
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shirin Strohmeier
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Temima Yellin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Philip S. Meade
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Disha Bhavsar
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Weina Sun
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lynda Coughlan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Center for Vaccine Development and Global Health (CVD), University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
96
|
Reynolds SL, Kaufman HW, Meyer WA, Bush C, Cohen O, Cronin K, Kabelac C, Leonard S, Anderson S, Petkov V, Lowy D, Sharpless N, Penberthy L. Risk of and duration of protection from SARS-CoV-2 reinfection assessed with real-world data. PLoS One 2023; 18:e0280584. [PMID: 36943829 PMCID: PMC10045607 DOI: 10.1371/journal.pone.0280584] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/03/2023] [Indexed: 03/23/2023] Open
Abstract
This retrospective observational study aimed to gain a better understanding of the protective duration of prior SARS-CoV-2 infection against reinfection. The objectives were two-fold: to assess the durability of immunity to SARS-CoV-2 reinfection among initially unvaccinated individuals with previous SARS-CoV-2 infection, and to evaluate the crude SARS-CoV-2 reinfection rate and associated risk factors. During the pandemic era time period from February 29, 2020, through April 30, 2021, 144,678,382 individuals with SARS-CoV-2 molecular diagnostic or antibody test results were studied. Rates of reinfection among index-positive individuals were compared to rates of infection among index-negative individuals. Factors associated with reinfection were evaluated using multivariable logistic regression. For both objectives, the outcome was a subsequent positive molecular diagnostic test result. Consistent with prior findings, the risk of reinfection among index-positive individuals was 87% lower than the risk of infection among index-negative individuals. The duration of protection against reinfection was stable over the median 5 months and up to 1-year follow-up interval. Factors associated with an increased reinfection risk included older age, comorbid immunologic conditions, and living in congregate care settings; healthcare workers had a decreased reinfection risk. This large US population-based study suggests that infection induced immunity is durable for variants circulating pre-Delta predominance.
Collapse
Affiliation(s)
- Shannon L. Reynolds
- Science and Delivery, Aetion, Inc., New York, New York, United States of America
| | - Harvey W. Kaufman
- Medical Affairs, Quest Diagnostics, Secaucus, New Jersey, United States of America
| | - William A. Meyer
- Medical Affairs, Quest Diagnostics, Secaucus, New Jersey, United States of America
| | - Chris Bush
- Science and Delivery, Aetion, Inc., New York, New York, United States of America
| | - Oren Cohen
- Office of the Chief Medical Office, Labcorp Drug Development, Burlington, North Carolina, United States of America
| | - Kathy Cronin
- Division of Cancer Control and Population Science, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Carly Kabelac
- Science and Delivery, Aetion, Inc., New York, New York, United States of America
| | - Sandy Leonard
- Parternships and Real World Data, HealthVerity, Philadelphia, PA, United States of America
| | - Steve Anderson
- Office of the Chief Medical Office, Labcorp Drug Development, Burlington, North Carolina, United States of America
| | - Valentina Petkov
- Division of Cancer Control and Population Science, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Douglas Lowy
- Division of Cancer Control and Population Science, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Norman Sharpless
- Division of Cancer Control and Population Science, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Lynne Penberthy
- Division of Cancer Control and Population Science, National Cancer Institute, Bethesda, Maryland, United States of America
| |
Collapse
|
97
|
Tuncer G, Geyiktepe-Guclu C, Surme S, Canel-Karakus E, Erdogan H, Bayramlar OF, Belge C, Karahasanoglu R, Copur B, Yazla M, Zerdali E, Nakir IY, Yildirim N, Kar B, Bozkurt M, Karanalbant K, Atasoy B, Takak H, Simsek-Yavuz S, Turkay R, M Sonmez M, Sengoz G, Pehlivanoglu F. Long-term effects of COVID-19 on lungs and the clinical relevance: a 6-month prospective cohort study. Future Microbiol 2023; 18:185-198. [PMID: 36916475 DOI: 10.2217/fmb-2022-0121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Background: We aimed to explore the prevalence of prolonged symptoms, pulmonary impairments and residual disease on chest tomography (CT) in COVID-19 patients at 6 months after acute illness. Methods: In this prospective, single-center study, hospitalized patients with radiologically and laboratory-confirmed COVID-19 were included. Results: A high proportion of the 116 patients reported persistent symptoms (n = 54; 46.6%). On follow-up CT, 33 patients (28.4%) demonstrated residual disease. Multivariate analyses revealed that only neutrophil-to-lymphocyte ratio was an independent predictor for residual disease. Conclusion: Hospitalized patients with mild/moderate COVID-19 still had persistent symptoms and were prone to develop long-term pulmonary sequelae on chest CT. However, it did not have a significant effect on long-term pulmonary functions.
Collapse
Affiliation(s)
- Gulsah Tuncer
- Department of Infectious Diseases & Clinical Microbiology, Haseki Training & Research Hospital, Istanbul, 34096, Turkey
| | - Ceyda Geyiktepe-Guclu
- Department of Infectious Diseases & Clinical Microbiology, Haseki Training & Research Hospital, Istanbul, 34096, Turkey
| | - Serkan Surme
- Department of Infectious Diseases & Clinical Microbiology, Haseki Training & Research Hospital, Istanbul, 34096, Turkey.,Department of Medical Microbiology, Institute of Graduate Studies, Istanbul University-Cerrahpasa, Istanbul, 34098, Turkey
| | - Evren Canel-Karakus
- Department of Pulmonary Medicine, Haseki Training & Research Hospital, Istanbul, 34096, Turkey
| | - Hatice Erdogan
- Department of Microbiology & Clinical Microbiology, Haseki Training & Research Hospital, Istanbul, 34096, Turkey
| | - Osman F Bayramlar
- Department of Public Health, Bakirkoy District Health Directorate, Istanbul, 34140, Turkey
| | - Cansu Belge
- Department of Radiology, Health Sciences University, Haseki Training & Research Hospital, Istanbul, 34096, Turkey
| | - Ridvan Karahasanoglu
- Department of Radiology, Health Sciences University, Haseki Training & Research Hospital, Istanbul, 34096, Turkey
| | - Betul Copur
- Department of Infectious Diseases & Clinical Microbiology, Haseki Training & Research Hospital, Istanbul, 34096, Turkey
| | - Meltem Yazla
- Department of Infectious Diseases & Clinical Microbiology, Haseki Training & Research Hospital, Istanbul, 34096, Turkey
| | - Esra Zerdali
- Department of Infectious Diseases & Clinical Microbiology, Haseki Training & Research Hospital, Istanbul, 34096, Turkey
| | - Inci Y Nakir
- Department of Infectious Diseases & Clinical Microbiology, Haseki Training & Research Hospital, Istanbul, 34096, Turkey
| | - Nihal Yildirim
- Department of Pulmonary Medicine, Haseki Training & Research Hospital, Istanbul, 34096, Turkey
| | - Bedriye Kar
- Department of Pulmonary Medicine, Haseki Training & Research Hospital, Istanbul, 34096, Turkey
| | - Mediha Bozkurt
- Department of Infectious Diseases & Clinical Microbiology, Haseki Training & Research Hospital, Istanbul, 34096, Turkey
| | - Kubra Karanalbant
- Department of Infectious Diseases & Clinical Microbiology, Haseki Training & Research Hospital, Istanbul, 34096, Turkey
| | - Burcu Atasoy
- Department of Infectious Diseases & Clinical Microbiology, Haseki Training & Research Hospital, Istanbul, 34096, Turkey
| | - Hindirin Takak
- Department of Infectious Diseases & Clinical Microbiology, Haseki Training & Research Hospital, Istanbul, 34096, Turkey
| | - Serap Simsek-Yavuz
- Department of Infectious Diseases & Clinical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, 34093, Turkey
| | - Rustu Turkay
- Department of Radiology, Health Sciences University, Haseki Training & Research Hospital, Istanbul, 34096, Turkey
| | - Mehmet M Sonmez
- Department of Orthopedic Surgery & Traumatology, Haseki Training & Research Hospital, Istanbul, 34096, Turkey
| | - Gonul Sengoz
- Department of Infectious Diseases & Clinical Microbiology, Haseki Training & Research Hospital, Istanbul, 34096, Turkey
| | - Filiz Pehlivanoglu
- Department of Infectious Diseases & Clinical Microbiology, Haseki Training & Research Hospital, Istanbul, 34096, Turkey
| |
Collapse
|
98
|
Gattinger P, Ohradanova-Repic A, Valenta R. Importance, Applications and Features of Assays Measuring SARS-CoV-2 Neutralizing Antibodies. Int J Mol Sci 2023; 24:ijms24065352. [PMID: 36982424 PMCID: PMC10048970 DOI: 10.3390/ijms24065352] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/03/2023] [Accepted: 03/05/2023] [Indexed: 03/17/2023] Open
Abstract
More than three years ago, the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) caused the unforeseen COVID-19 pandemic with millions of deaths. In the meantime, SARS-CoV-2 has become endemic and is now part of the repertoire of viruses causing seasonal severe respiratory infections. Due to several factors, among them the development of SARS-CoV-2 immunity through natural infection, vaccination and the current dominance of seemingly less pathogenic strains belonging to the omicron lineage, the COVID-19 situation has stabilized. However, several challenges remain and the possible new occurrence of highly pathogenic variants remains a threat. Here we review the development, features and importance of assays measuring SARS-CoV-2 neutralizing antibodies (NAbs). In particular we focus on in vitro infection assays and molecular interaction assays studying the binding of the receptor binding domain (RBD) with its cognate cellular receptor ACE2. These assays, but not the measurement of SARS-CoV-2-specific antibodies per se, can inform us of whether antibodies produced by convalescent or vaccinated subjects may protect against the infection and thus have the potential to predict the risk of becoming newly infected. This information is extremely important given the fact that a considerable number of subjects, in particular vulnerable persons, respond poorly to the vaccination with the production of neutralizing antibodies. Furthermore, these assays allow to determine and evaluate the virus-neutralizing capacity of antibodies induced by vaccines and administration of plasma-, immunoglobulin preparations, monoclonal antibodies, ACE2 variants or synthetic compounds to be used for therapy of COVID-19 and assist in the preclinical evaluation of vaccines. Both types of assays can be relatively quickly adapted to newly emerging virus variants to inform us about the magnitude of cross-neutralization, which may even allow us to estimate the risk of becoming infected by newly appearing virus variants. Given the paramount importance of the infection and interaction assays we discuss their specific features, possible advantages and disadvantages, technical aspects and not yet fully resolved issues, such as cut-off levels predicting the degree of in vivo protection.
Collapse
Affiliation(s)
- Pia Gattinger
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Anna Ohradanova-Repic
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Rudolf Valenta
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- Karl Landsteiner University, 3500 Krems an der Donau, Austria
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, 119435 Moscow, Russia
- NRC Institute of Immunology FMBA of Russia, 115478 Moscow, Russia
- Correspondence:
| |
Collapse
|
99
|
Rana R, Kant R, Kumra T, Gupta S, Rana DS, Ganguly NK. An update on SARS-CoV-2 immunization and future directions. Front Pharmacol 2023; 14:1125305. [PMID: 36969857 PMCID: PMC10033701 DOI: 10.3389/fphar.2023.1125305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/09/2023] [Indexed: 03/29/2023] Open
Abstract
Millions of people have died as a result of SARS-CoV-2, which was first discovered in China and has since spread globally. Patients with SARS-CoV-2 infection may show a range of symptoms, including fever, coughing, and shortness of breath, or they may show no symptoms at all. To treat COVID-19 symptoms and avoid serious infections, many medications and vaccinations have been employed. However, to entirely eradicate COVID-19 from the world, next-generation vaccine research is required because of the devastating consequences it is having for humanity and every nation's economy. Scientists are working hard to eradicate this dangerous virus across the world. SARS-CoV-2 has also undergone significant mutation, leading to distinct viral types such as the alpha, beta, gamma, delta, and omicron variants. This has sparked discussion about the effectiveness of current vaccines for the newly formed variants. A proper comparison of these vaccinations is required to compare their efficacy as the number of people immunized against SARS-CoV-2 globally increases. Population-level statistics evaluating the capacity of these vaccines to reduce infection are therefore being developed. In this paper, we analyze the many vaccines on the market in terms of their production process, price, dosage needed, and efficacy. This article also discusses the challenges of achieving herd immunity, the likelihood of reinfection, and the importance of convalescent plasma therapy in reducing infection.
Collapse
Affiliation(s)
- Rashmi Rana
- Department of Research, Sir Ganga Ram Hospital, New Delhi, India
| | - Ravi Kant
- Department of Research, Sir Ganga Ram Hospital, New Delhi, India
| | - Tanya Kumra
- Department of Research, Sir Ganga Ram Hospital, New Delhi, India
| | - Sneha Gupta
- Department of Research, Sir Ganga Ram Hospital, New Delhi, India
| | | | | |
Collapse
|
100
|
Tolan NV, DeSimone MS, Fernandes MD, Lewis JE, Simmons DP, Schur PH, Brigl M, Tanasijevic MJ, Desjardins M, Sherman AC, Baden LR, Snyder M, Melanson SE. Lessons learned: A look back at the performance of nine COVID-19 serologic assays and their proposed utility. Clin Biochem 2023; 117:60-68. [PMID: 36878344 PMCID: PMC9985916 DOI: 10.1016/j.clinbiochem.2023.03.003] [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: 10/11/2022] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/07/2023]
Abstract
BACKGROUND Serologic assays for the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been proposed to assist with the acute diagnosis of infection, support epidemiological studies, identify convalescent plasma donors, and evaluate vaccine response. METHODS We report an evaluation of nine serologic assays: Abbott (AB) and Epitope (EP) IgG and IgM, EUROIMMUN (EU) IgG and IgA, Roche anti-N (RN TOT) and anti-S (RS TOT) total antibody, and DiaSorin (DS) IgG. We evaluated 291 negative controls (NEG CTRL), 91 PCR positive (PCR POS) patients (179 samples), 126 convalescent plasma donors (CPD), 27 healthy vaccinated donors (VD), and 20 allogeneic hematopoietic stem cell transplant (HSCT) recipients (45 samples). RESULTS We observed good agreement with the method performance claims for specificity (93-100%) in NEG CTRL but only 85% for EU IgA. The sensitivity claims in the first 2 weeks of symptom onset was lower (26-61%) than performance claims based on > 2 weeks since PCR positivity. We observed high sensitivities (94-100%) in CPD except for AB IgM (77%), EP IgM (0%). Significantly higher RS TOT was observed for Moderna vaccine recipients then Pfizer (p-values < 0.0001). A sustained RS TOT response was observed for the five months following vaccination. HSCT recipients demonstrated significantly lower RS TOT than healthy VD (p < 0.0001) at dose 2 and 4 weeks after. CONCLUSIONS Our data suggests against the use of anti-SARS-CoV-2 assays to aid in acute diagnosis. RN TOT and RS TOT can readily identify past-resolved infection and vaccine response in the absence of native infection. We provide an estimate of expected antibody response in healthy VD over the time course of vaccination for which to compare antibody responses in immunosuppressed patients.
Collapse
Affiliation(s)
- Nicole V Tolan
- Brigham and Women's Hospital, Department of Pathology, Boston, MA, United States; Harvard Medical School, Boston, MA, United States.
| | - Mia S DeSimone
- Brigham and Women's Hospital, Department of Pathology, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Maria D Fernandes
- Brigham and Women's Hospital, Department of Pathology, Boston, MA, United States
| | - Joshua E Lewis
- Brigham and Women's Hospital, Department of Pathology, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Daimon P Simmons
- Brigham and Women's Hospital, Department of Pathology, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Peter H Schur
- Brigham and Women's Hospital, Department of Pathology, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Manfred Brigl
- Brigham and Women's Hospital, Department of Pathology, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Milenko J Tanasijevic
- Brigham and Women's Hospital, Department of Pathology, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Michaël Desjardins
- Harvard Medical School, Boston, MA, United States; Brigham and Women's Hospital, Department of Medicine, Division of Infectious Diseases, Boston, MA, United States
| | - Amy C Sherman
- Harvard Medical School, Boston, MA, United States; Brigham and Women's Hospital, Department of Medicine, Division of Infectious Diseases, Boston, MA, United States
| | - Lindsey R Baden
- Harvard Medical School, Boston, MA, United States; Brigham and Women's Hospital, Department of Medicine, Division of Infectious Diseases, Boston, MA, United States
| | | | - Stacy Ef Melanson
- Brigham and Women's Hospital, Department of Pathology, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| |
Collapse
|