1
|
Oliva-Ariza G, Criado I, Fuentes-Herrero B, Carbonell C, Sánchez-Gallego JI, López-Bernús A, Gutiérrez ML, Rolo-Ramírez A, Bernal-Ribes M, Almenara-Morales Y, Lecrevisse Q, van Dongen JJM, Marcos M, Almeida J, Orfao A. Early Immune Cell and Antibody Kinetics Following SARS-CoV-2 Vaccination in Healthy Adults and Low-Count Monoclonal B-Cell Lymphocytosis. Int J Mol Sci 2025; 26:681. [PMID: 39859394 PMCID: PMC11765611 DOI: 10.3390/ijms26020681] [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: 12/09/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
The early immune kinetics after SARS-CoV-2 vaccination remain poorly understood, particularly among individuals with low-count monoclonal B-cell lymphocytosis (MBLlo). We investigated the cellular and humoral kinetics in the blood of 50 non-MBL healthy donors (HD) vs. 16 MBLlo subjects after SARS-CoV-2 vaccination, who were subclassified according to their history of previous exposure to SARS-CoV-2 into SARS-CoV-2 naïve and previously infected subjects. Overall, we found decreased neutrophil and lymphocyte counts at day +4 following each dose in non-MBL HD, together with an earlier and higher increase in plasma cell (PC) counts and SARS-CoV-2-specific antibody levels after the first vaccine in previously infected non-MBL HD. MBLlo subjects showed a similar profile, except for lower B-cell and higher PC counts after vaccination, and a trend towards a higher (but delayed) antibody response. In summary, we found different cell-kinetic profiles following vaccination in SARS-CoV-2 naïve vs. previously infected non-MBL HD (earlier PC and antibody responses in the latter group); additionally, MBLlo subjects had significantly lower B-cell and higher PC counts after vaccination, and a delayed SARS-CoV-2-specific antibody response.
Collapse
Affiliation(s)
- Guillermo Oliva-Ariza
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Ignacio Criado
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Blanca Fuentes-Herrero
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Cristina Carbonell
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Department of Internal Medicine, University Hospital of Salamanca, 37007 Salamanca, Spain; (A.R.-R.); (Y.A.-M.)
- Department of Infectious Diseases, University Hospital of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), 37007 Salamanca, Spain
| | - José Ignacio Sánchez-Gallego
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Amparo López-Bernús
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Department of Internal Medicine, University Hospital of Salamanca, 37007 Salamanca, Spain; (A.R.-R.); (Y.A.-M.)
- Department of Infectious Diseases, University Hospital of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), 37007 Salamanca, Spain
| | - María Laura Gutiérrez
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alejandro Rolo-Ramírez
- Department of Internal Medicine, University Hospital of Salamanca, 37007 Salamanca, Spain; (A.R.-R.); (Y.A.-M.)
| | - Marta Bernal-Ribes
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Yolimar Almenara-Morales
- Department of Internal Medicine, University Hospital of Salamanca, 37007 Salamanca, Spain; (A.R.-R.); (Y.A.-M.)
| | - Quentin Lecrevisse
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jacques J. M. van Dongen
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
| | - Miguel Marcos
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Department of Internal Medicine, University Hospital of Salamanca, 37007 Salamanca, Spain; (A.R.-R.); (Y.A.-M.)
| | - Julia Almeida
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alberto Orfao
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
2
|
de Oliveira LA, de Morais IRB, Marchioro SB, de Almeida GB, de Almeida de Souza GH, da Silva Ferreira T, Rossoni R, de Oliveira Barbosa D, Navarini VJ, Croda J, Torres AJL, Simionatto S. Assessment of the BNT162B2 COVID-19 vaccine immune response in Brazilian indigenous adolescents. Vaccine 2025; 43:126494. [PMID: 39571350 DOI: 10.1016/j.vaccine.2024.126494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/21/2024] [Accepted: 10/25/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND COVID-19 vaccination of minors is crucial for global pandemic control, especially among indigenous populations, who are often more vulnerable due to limited healthcare resources and communal living settings. OBJECTIVES To assess the immunogenicity responses of the BNT162b2 vaccine in immunized Brazilian indigenous adolescents. METHODS A cohort study was conducted with indigenous adolescents aged 12 to 18 years residing in the largest peri-urban indigenous region in Brazil. SARS-CoV-2-specific immune responses were analyzed before (D1) and after (D2) completion of the vaccination schedule. Demographic data were collected using a questionnaire. RESULTS Of the 129 adolescents invited, 98 (75.96 %) participated in the study. Most were of Guarani ethnicity, single, had lower incomes, and were educated only to the elementary level. Post-vaccination, a statistically significant increase was noted in IgG concentration (24.03 % to 37.02 %). Increases were observed in B lymphocytes (11.88 to 13.92 cells/mm3), memory B cells (13.58 to 15.96 cells/mm3), NK cells (20.23 to 24.08 cells/mm3), and non-classical monocytes (9.23 to 11.34 cells/mm3), while CD8+ T cells decreased (24.41 to 21.69 cells/mm3). Adolescents with prior exposure to the virus showed increased levels of B lymphocytes and CD8+ T cells. No significant changes were observed in other cell subpopulations from exposure to the virus. CONCLUSION Elevated levels of antibodies and certain cell subpopulations were observed in vaccinated adolescents, confirming the effectiveness of the BNT162b2 vaccine in maintaining humoral and cellular responses. This study is the first to describe data from indigenous minors vaccinated against COVID-19 with the BNT162b2 vaccine, highlighting the importance of vaccination efforts and the potential need for booster doses.
Collapse
Affiliation(s)
- Laís Albuquerque de Oliveira
- Health Science Research Laboratory, Federal University of Grande Dourados, Dourados, Mato Grosso, do Sul, Brazil
| | - Isa Rita Brito de Morais
- Health Science Research Laboratory, Federal University of Grande Dourados, Dourados, Mato Grosso, do Sul, Brazil
| | - Silvana Beutinger Marchioro
- Laboratory of Immunology and Molecular Biology, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Gabriel Barroso de Almeida
- Laboratory of Immunology and Molecular Biology, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, Brazil
| | | | - Tiago da Silva Ferreira
- Health Science Research Laboratory, Federal University of Grande Dourados, Dourados, Mato Grosso, do Sul, Brazil
| | - Regina Rossoni
- Health Science Research Laboratory, Federal University of Grande Dourados, Dourados, Mato Grosso, do Sul, Brazil
| | - Dyjaene de Oliveira Barbosa
- Laboratory of Immunology and Molecular Biology, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Vinicius João Navarini
- University Hospital of the Federal University of Grande Dourados - Brazilian Hospital Services Company EBSERH, Dourados, Mato Grosso, do Sul, Brazil
| | - Julio Croda
- School of Medicine, Federal University of Mato Grosso do Sul, Campo Grande, Brazil.; Oswaldo Cruz Foundation, Campo Grande, Brazil.; Department of Epidemiology of Microbial Diseases, Yale University School of Public Health, New Haven, CT, United States of America
| | - Alex José Leite Torres
- Laboratory of Immunology and Molecular Biology, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Simone Simionatto
- Health Science Research Laboratory, Federal University of Grande Dourados, Dourados, Mato Grosso, do Sul, Brazil..
| |
Collapse
|
3
|
Rothoeft T, Maier C, Talarico A, Hoffmann A, Schlegtendal A, Lange B, Petersmann A, Denz R, Timmesfeld N, Toepfner N, Vidal-Blanco E, Pfaender S, Lücke T, Brinkmann F. Natural and hybrid immunity after SARS-CoV-2 infection in children and adolescents. Infection 2024; 52:1449-1458. [PMID: 38499828 PMCID: PMC11288991 DOI: 10.1007/s15010-024-02225-w] [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: 10/07/2023] [Accepted: 02/24/2024] [Indexed: 03/20/2024]
Abstract
PURPOSE In contrast to adults, immune protection against SARS-CoV-2 in children and adolescents with natural or hybrid immunity is still poorly understood. The aim of this study was to analyze different immune compartments in different age groups and whether humoral immune reactions correlate with a cellular immune response. METHODS 72 children and adolescents with a preceding SARS-CoV-2 infection were recruited. 37 were vaccinated with an RNA vaccine (BNT162b2). Humoral immunity was analyzed 3-26 months (median 10 months) after infection by measuring Spike protein (S), nucleocapsid (NCP), and neutralizing antibodies (nAB). Cellular immunity was analyzed using a SARS-CoV-2-specific interferon-γ release assay (IGRA). RESULTS All children and adolescents had S antibodies; titers were higher in those with hybrid immunity (14,900 BAU/ml vs. 2118 BAU/ml). NCP antibodies were detectable in > 90%. Neutralizing antibodies (nAB) were more frequently detected (90%) with higher titers (1914 RLU) in adolescents with hybrid immunity than in children with natural immunity (62.5%, 476 RLU). Children with natural immunity were less likely to have reactive IGRAs (43.8%) than adolescents with hybrid immunity (85%). The amount of interferon-γ released by T cells was comparable in natural and hybrid immunity. CONCLUSION Spike antibodies are the most reliable markers to monitor an immune reaction against SARS-CoV-2. High antibody titers of spike antibodies and nAB correlated with cellular immunity, a phenomenon found only in adolescents with hybrid immunity. Hybrid immunity is associated with markedly higher antibody titers and a higher probability of a cellular immune response than a natural immunity.
Collapse
Affiliation(s)
- T Rothoeft
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University, Bochum, Germany.
| | - C Maier
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - A Talarico
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - A Hoffmann
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - A Schlegtendal
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - B Lange
- Department of Epidemiology, Helmholtz Centre for Infection Research, Brunswick, Germany
| | - A Petersmann
- University Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Oldenburg, Oldenburg, Germany
- University Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - R Denz
- Department of Medical Informatics, Biometry and Epidemiology, Ruhr-University Bochum, Bochum, Germany
| | - N Timmesfeld
- Department of Medical Informatics, Biometry and Epidemiology, Ruhr-University Bochum, Bochum, Germany
| | - N Toepfner
- Department of Pediatrics, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - E Vidal-Blanco
- Department of Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - S Pfaender
- Department of Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - T Lücke
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - F Brinkmann
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University, Bochum, Germany
- University Children's Hospital, Lübeck, Germany
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Lübeck, Germany
| |
Collapse
|
4
|
Tani N, Ikematsu H, Watanabe H, Goto T, Yanagihara Y, Kurata Y, Harada Y, Horiuchi T, Akashi K, Shimono N, Chong Y. Reduction of adverse reactions and correlation between post-vaccination fever and specific antibody response across successive SARS-CoV-2 mRNA vaccinations. Vaccine X 2024; 18:100489. [PMID: 38699157 PMCID: PMC11063515 DOI: 10.1016/j.jvacx.2024.100489] [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: 02/28/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024] Open
Abstract
Background SARS-CoV-2 mRNA vaccination, recognized for high immunogenicity, frequently induces adverse reactions, especially fever. We previously reported a correlation between post-vaccination fever and specific antibody responses to the primary series and first booster. We herein report changes in adverse reactions and the correlation between post-vaccination fever and antibody responses across successive vaccinations, from monovalent to bivalent mRNA vaccines. Methods This cohort study was conducted at a Japanese hospital to investigate adverse reactions to the monovalent primary, first booster, and BA.4/5 bivalent BNT162b2 vaccinations. Local and systemic reactions were reported through a self-reporting diary after each dose. The spike-specific IgG titers were measured following each vaccination. Results Across 727 vaccinations in the vaccine series, the bivalent booster induced fewer adverse reactions than earlier doses. Fever ≥ 38.0 °C was significantly less frequent in the bivalent booster (12.3 %) compared to the primary series and monovalent booster (22.0 %, 26.2 %, p < 0.001). Reaction severity was also reduced in the bivalent booster. In the analysis of 70 participants with complete data for all doses, post-vaccination fever ≥ 38.0 °C exhibited the highest relative risk (RR) among all solicited reactions throughout the vaccine series (RR: 5.24 [95 % CI: 2.40-11.42] for monovalent and 6.24 [95 % CI: 2.14-18.15] for bivalent). The frequency of fever ≥ 38.0 °C after all doses was 8.6 % (6/70), with no fever ≥ 39.0 °C across all vaccinations. A high-grade post-vaccination fever was correlated with higher IgG titers, with multivariate analyses confirming this correlation as independent for each dose and unaffected by previous post-vaccination fever. Conclusions The bivalent mRNA vaccine booster showed fewer and milder adverse reactions than the monovalent doses. Although vaccinees with a history of post-vaccination fever were more likely to experience fever after a subsequent dose, such recurrences were infrequent. A correlation between post-vaccination fever and increased IgG titers was identified for each vaccination, irrespective of post-vaccination fever history.
Collapse
Affiliation(s)
- Naoki Tani
- Department of Infectious Diseases, Fukuoka City Hospital, Fukuoka, Japan
| | | | - Haruka Watanabe
- Department of Clinical Immunology, Rheumatology, and Infectious Disease, Kyushu University Hospital, Fukuoka, Japan
| | - Takeyuki Goto
- Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | | | | | - Yukiko Harada
- Department of Infectious Diseases, Fukuoka City Hospital, Fukuoka, Japan
| | | | - Koichi Akashi
- Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Nobuyuki Shimono
- Center for the Study of Global Infection, Kyushu University Hospital, Fukuoka, Japan
| | - Yong Chong
- Department of Clinical Immunology, Rheumatology, and Infectious Disease, Kyushu University Hospital, Fukuoka, Japan
| |
Collapse
|
5
|
Montmaneix-Engels F, Dimeglio C, Staes L, Da Silva I, Porcheron M, Jougla I, Hérin F, Izopet J. Study of the cellular and humoral immune responses to SARS-CoV-2 vaccination. Heliyon 2024; 10:e29116. [PMID: 38601689 PMCID: PMC11004869 DOI: 10.1016/j.heliyon.2024.e29116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 04/12/2024] Open
Abstract
Our understanding of cellular immunity in response to COVID-19 infection or vaccination is limited because of less commonly used techniques. We investigated both the cellular and humoral immune responses before and after the administration of a third dose of the SARS-CoV-2 vaccine among a group of healthcare workers. Cellular immunity was evaluated using the VIDAS interferon-gamma (IFNγ) RUO test, which enables automated measurement of IFNγ levels after stimulating peripheral blood lymphocytes. Booster doses significantly enhanced both cellular and humoral immunity. Concerning cellular response, the booster dose increased the percentage of positive IFNγ release assay (IGRA) results but no difference in IFNγ release was found. The cellular response was not associated with protection against SARS-CoV-2 infection. Interestingly, vaccinated and infected healthcare workers exhibited the highest levels of anti-spike and neutralizing antibodies. In conclusion, the IGRA is a simple method for measuring cellular immune responses after vaccination. However, its usefulness as a complement to the study of humoral responses is yet to be demonstrated in future research.
Collapse
Affiliation(s)
- Faustine Montmaneix-Engels
- INSERM UMR1291-CNRS UMR5051-University Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, 31300, Toulouse, France
- Toulouse III Paul Sabatier University, 31062, Toulouse, France
| | - Chloé Dimeglio
- INSERM UMR1291-CNRS UMR5051-University Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, 31300, Toulouse, France
- CHU Toulouse, Purpan Hospital, Virology Laboratory, 31300, Toulouse, France
| | - Laeticia Staes
- CHU Toulouse, Purpan Hospital, Virology Laboratory, 31300, Toulouse, France
| | - Isabelle Da Silva
- CHU Toulouse, Purpan Hospital, Virology Laboratory, 31300, Toulouse, France
| | - Marion Porcheron
- CHU Toulouse, Purpan Hospital, Virology Laboratory, 31300, Toulouse, France
| | - Isabelle Jougla
- Occupational Diseases Department, Toulouse University Hospital, 31000, Toulouse, France
| | - Fabrice Hérin
- Occupational Diseases Department, Toulouse University Hospital, 31000, Toulouse, France
- UMR1295, Joint Research Unit INSERM- University Toulouse III Paul Sabatier, Centre for Epidemiology and Research in Population Health Unit (CERPOP), 31000, Toulouse, France
| | - Jacques Izopet
- INSERM UMR1291-CNRS UMR5051-University Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, 31300, Toulouse, France
- Toulouse III Paul Sabatier University, 31062, Toulouse, France
- CHU Toulouse, Purpan Hospital, Virology Laboratory, 31300, Toulouse, France
| |
Collapse
|
6
|
Lemieux A, Sannier G, Nicolas A, Nayrac M, Delgado GG, Cloutier R, Brassard N, Laporte M, Duchesne M, Sreng Flores AM, Finzi A, Tastet O, Dubé M, Kaufmann DE. Enhanced detection of antigen-specific T cells by a multiplexed AIM assay. CELL REPORTS METHODS 2024; 4:100690. [PMID: 38228152 PMCID: PMC10831934 DOI: 10.1016/j.crmeth.2023.100690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 11/21/2023] [Accepted: 12/18/2023] [Indexed: 01/18/2024]
Abstract
Broadly applicable methods to identify and characterize antigen-specific CD4+ and CD8+ T cells are key to immunology research, including studies of vaccine responses and immunity to infectious diseases. We developed a multiplexed activation-induced marker (AIM) assay that presents several advantages compared to single pairs of AIMs. The simultaneous measurement of four AIMs (CD69, 4-1BB, OX40, and CD40L) creates six AIM pairs that define CD4+ T cell populations with partial and variable overlap. When combined in an AND/OR Boolean gating strategy for analysis, this approach enhances CD4+ T cell detection compared to any single AIM pair, while CD8+ T cells are dominated by CD69/4-1BB co-expression. Supervised and unsupervised clustering analyses show differential expression of the AIMs in defined T helper lineages and that multiplexing mitigates phenotypic biases. Paired and unpaired comparisons of responses to infections (HIV and cytomegalovirus [CMV]) and vaccination (severe acute respiratory syndrome coronavirus 2 [SARS-CoV-2]) validate the robustness and versatility of the method.
Collapse
Affiliation(s)
- Audrée Lemieux
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Gérémy Sannier
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Alexandre Nicolas
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Manon Nayrac
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | | | - Rose Cloutier
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada
| | | | | | | | | | - Andrés Finzi
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Olivier Tastet
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada
| | - Mathieu Dubé
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada.
| | - Daniel E Kaufmann
- Centre de Recherche du CHUM, Montreal, QC H2X 0A9, Canada; Consortium for HIV/AIDS Vaccine Development (CHAVD), La Jolla, CA, USA; Département de Médecine, Université de Montréal, Montreal, QC H2X 0A9, Canada; Division of Infectious Diseases, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
7
|
Arientová S, Matúšková K, Bartoš O, Holub M, Beran O. Specific immune responses after BNT162b2 mRNA vaccination and COVID-19 infection. Front Immunol 2023; 14:1271353. [PMID: 37920457 PMCID: PMC10619853 DOI: 10.3389/fimmu.2023.1271353] [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: 08/02/2023] [Accepted: 09/28/2023] [Indexed: 11/04/2023] Open
Abstract
Although vaccines against COVID-19 are effective tools in preventing severe disease, recent studies have shown enhanced protection after vaccine boosters. The aim of our study was to examine the dynamics and duration of both humoral and cellular immune responses following a three-dose regimen of the BNT162b2 mRNA vaccine. In a longitudinal prospective study we enrolled 86 adults who received the BNT162b2 vaccine, 35 unvaccinated individuals with a history of mild COVID-19 and a control group of 30 healthy SARS-CoV-2 seronegative persons. We assessed the SARS-CoV-2-specific T cell responses and IgG production up to 12 months post the third BNT162b2 dose in 24 subjects. The vaccinated group had significantly higher IgG antibody levels after two doses compared to the convalescent group (p<0.001). After the third dose, IgG levels surged beyond those detected after the second dose (p<0.001). Notably, these elevated IgG levels were maintained 12 months post the third dose. After two doses, specific T cell responses were detected in 87.5% of the vaccinated group. Additionally, there was a significant decrease before the third dose. However, post the third dose, specific T cell responses surged and remained stable up to the 12-month period. Our findings indicate that the BNT162b2 vaccine induces potent and enduring humoral and cellular responses, which are notably enhanced by the third dose and remain persistant without a significant decline a year after the booster. Further research is essential to understand the potential need for subsequent boosters.
Collapse
Affiliation(s)
- Simona Arientová
- Department of Infectious Diseases, First Faculty of Medicine, Charles University and Military University Hospital Prague, Prague, Czechia
| | - Kateřina Matúšková
- Department of Infectious Diseases, First Faculty of Medicine, Charles University and Military University Hospital Prague, Prague, Czechia
| | - Oldřich Bartoš
- Military Health Institute, Military Medical Agency, Prague, Czechia
| | - Michal Holub
- Department of Infectious Diseases, First Faculty of Medicine, Charles University and Military University Hospital Prague, Prague, Czechia
| | - Ondřej Beran
- Department of Infectious Diseases, First Faculty of Medicine, Charles University and Military University Hospital Prague, Prague, Czechia
| |
Collapse
|
8
|
Lamara Mahammed L, Bensaid K, Ait-Seddik S, Larinouna A, Brahimi G, Belkaid R, Hamzaoui O, Rouaki SM, Idder C, Allam I, Djidjik R. Improved Performance of the QuantiFERON-SARS-CoV-2 Assay with the Extended Set. Viruses 2023; 15:v15051179. [PMID: 37243265 DOI: 10.3390/v15051179] [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: 04/17/2023] [Revised: 05/09/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Multiple assays have been developed for the characterization of the functional activation of SARS-CoV-2 specific T-cells. This study was conducted to assess the post-vaccination and post-infection T cell response, as detected by the QuantiFERON-SARS-CoV-2 assay using the combination of three SARS-CoV-2 specific antigens (Ag1, Ag2 and Ag3). An amount of 75 participants with different infection and vaccination backgrounds were recruited for the evaluation of humoral and cellular immune responses. An elevated IFN-γ response in at least one Ag tube was observed in 69.2% of convalescent subjects and 63.9% of vaccinated ones. Interestingly, in a healthy unvaccinated case and three convalescents with negative IgG-RBD, we detected a positive QuantiFERON test after stimulation with Ag3. The majority of the T cell responders reacted simultaneously to the three SARS-CoV-2 specific antigens, and Ag3 demonstrated the highest rate of reactivity. At univariable analysis, the only factor that was associated with an absence of a cellular response was time from blood collection, being less than 30 days (OR:3.5, CI95% [1.15-10.50], p = 0.028). Overall, the inclusion of Ag3 improved the performance of the QuantiFERON-SARS-CoV-2 and showed a particular interest among subjects who fail to achieve a measurable antibody response after infection or vaccination.
Collapse
Affiliation(s)
- Lydia Lamara Mahammed
- Immunology Department, Beni-Messous Teaching Hospital, Faculty of Pharmacy, University of Algiers, Algiers 16000, Algeria
| | - Kahina Bensaid
- Immunology Department, Beni-Messous Teaching Hospital, Faculty of Pharmacy, University of Algiers, Algiers 16000, Algeria
| | - Sarah Ait-Seddik
- Epidemiology Department, Beni-Messous Teaching Hospital, Faculty of Medicine, University of Algiers, Algiers 16000, Algeria
| | - Amel Larinouna
- Epidemiology Department, Beni-Messous Teaching Hospital, Faculty of Medicine, University of Algiers, Algiers 16000, Algeria
| | - Ghania Brahimi
- Epidemiology Department, Beni-Messous Teaching Hospital, Faculty of Medicine, University of Algiers, Algiers 16000, Algeria
| | - Rosa Belkaid
- Epidemiology Department, Beni-Messous Teaching Hospital, Faculty of Medicine, University of Algiers, Algiers 16000, Algeria
| | | | - Soumia Meriem Rouaki
- Occupational Medicine, Beni-Messous Teaching Hospital, Faculty of Medicine, University of Algiers, Algiers 16000, Algeria
| | - Cherifa Idder
- Occupational Medicine, Beni-Messous Teaching Hospital, Faculty of Medicine, University of Algiers, Algiers 16000, Algeria
| | - Ines Allam
- Immunology Department, Beni-Messous Teaching Hospital, Faculty of Pharmacy, University of Algiers, Algiers 16000, Algeria
| | - Reda Djidjik
- Immunology Department, Beni-Messous Teaching Hospital, Faculty of Pharmacy, University of Algiers, Algiers 16000, Algeria
| |
Collapse
|
9
|
Roškar Z, Dreisinger M, Tič P, Homšak E, Bevc S, Goropevšek A. New Flow Cytometric Methods for Monitoring STAT5 Signaling Reveal Responses to SARS-CoV-2 Antigen-Specific Stimulation in FOXP3+ Regulatory T Cells also in Patients with Advanced Chronic Lymphocytic Leukemia. BIOSENSORS 2023; 13:bios13050539. [PMID: 37232900 DOI: 10.3390/bios13050539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/04/2023] [Accepted: 05/08/2023] [Indexed: 05/27/2023]
Abstract
Increased frequency of CD4+CD25+ regulatory T-cells (Treg) has been associated with disease progression in chronic lymphocytic leukemia (CLL). Flow cytometric methods, which allow for the simultaneous analysis of their specific transcription factor Foxp3 and activated STAT proteins, together with proliferation can help to elucidate the signaling mechanisms driving Treg expansion and suppression of FOXP3- conventional CD4+T-cells (Tcon). Herein, we first report a novel approach in which STAT5 phosphorylation (pSTAT5) and proliferation (BrdU-FITC incorporation) could be analyzed specifically in FOXP3+ and FOXP3- responding cells after CD3/CD28 stimulation. The addition of magnetically purified CD4+CD25+ T-cells from healthy donors to cocultured autologous CD4+CD25- T-cells resulted in suppression of Tcon cell cycle progression accompanied by a decrease in pSTAT5. Next, a method using imaging flow cytometry is presented for the detection of cytokine-dependent pSTAT5 nuclear translocation in FOXP3-expressing cells. Finally, we discuss our experimental data obtained by combining Treg pSTAT5 analysis and antigen-specific stimulation with SARS-CoV-2 antigens. Applying these methods on samples from patients revealed Treg responses to antigen-specific stimulation and significantly higher basal pSTAT5 in CLL patients treated with immunochemotherapy. Thus, we speculate that through the use of this pharmacodynamic tool, the efficacy of immunosuppressive drugs and their possible off-target effects can be assessed.
Collapse
Affiliation(s)
- Zlatko Roškar
- Department of Haematology, University Medical Centre Maribor, 2000 Maribor, Slovenia
| | - Mojca Dreisinger
- Department of Haematology, University Medical Centre Maribor, 2000 Maribor, Slovenia
| | - Primož Tič
- Department of Laboratory Diagnostics, University Medical Centre Maribor, 2000 Maribor, Slovenia
| | - Evgenija Homšak
- Department of Laboratory Diagnostics, University Medical Centre Maribor, 2000 Maribor, Slovenia
| | - Sebastjan Bevc
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
- Department of Nephrology, University Medical Center Maribor, 2000 Maribor, Slovenia
| | - Aleš Goropevšek
- Department of Laboratory Diagnostics, University Medical Centre Maribor, 2000 Maribor, Slovenia
| |
Collapse
|
10
|
Lee TH, Nam M, Seo JD, Kim H, Kim HR, Hur M, Yun YM, Moon HW. Evaluation of Cellular Responses to ChAdOx1-nCoV-19 and BNT162b2 Vaccinations. Ann Lab Med 2023; 43:290-294. [PMID: 36544341 PMCID: PMC9791012 DOI: 10.3343/alm.2023.43.3.290] [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: 06/14/2022] [Revised: 09/22/2022] [Accepted: 10/24/2022] [Indexed: 12/24/2022] Open
Abstract
While numerous studies have evaluated humoral responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines, data on the cellular responses to these vaccines remain sparse. We evaluated T cell responses to ChAdOx1-nCoV-19 and BNT162b2 vaccinations using an interferon gamma (IFN-γ) release assay (IGRA). ChAdOx1-nCoV-19- and BNT162b2-vaccinated participants initially showed stronger T cell responses than unvaccinated controls. The T cell response decreased over time and increased substantially after the administration of a BNT162b2 booster dose. Changes in the T cell response were less significant than those in the anti-receptor-binding domain IgG antibody titer. The study results can serve as baseline data for T cell responses after SARS-CoV-2 vaccination and suggest that the IGRA can be useful in monitoring immunogenicity.
Collapse
Affiliation(s)
- Tae Hwan Lee
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul, Korea
| | - Minjeong Nam
- Department of Laboratory Medicine, Korea University Anam Hospital, Seoul, Korea
| | - Jong Do Seo
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul, Korea
| | - Hanah Kim
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul, Korea
| | - Hae-Rim Kim
- Department of Internal Medicine, Konkuk University School of Medicine, Seoul, Korea
| | - Mina Hur
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul, Korea
| | - Yeo-Min Yun
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul, Korea
| | - Hee-Won Moon
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul, Korea,Corresponding author: Hee-Won Moon, M.D. Department of Laboratory Medicine, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Korea Tel: +82-2-2030-5583 Fax: +82-2-2030-5587 E-mail:
| |
Collapse
|
11
|
Evaluation of T cell responses with the QuantiFERON SARS-CoV-2 assay in individuals with 3 doses of BNT162b2 vaccine, SARS-CoV-2 infection, or hybrid immunity. Diagn Microbiol Infect Dis 2023; 106:115948. [PMID: 37094435 PMCID: PMC10060202 DOI: 10.1016/j.diagmicrobio.2023.115948] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/14/2023] [Accepted: 03/26/2023] [Indexed: 04/01/2023]
Abstract
Cellular immunity after SARS-CoV-2 infection or immunization may be important for long-lasting protection against severe COVID-19 disease. We investigated cellular immune responses after SARS-CoV-2 infection and/or vaccination with an interferon-γ release assay (QuantiFERON, QFN), in parallel, with humoral immunity assessment. We recruited 41 participants: unvaccinated convalescent children and adults and vaccinated uninfected or vaccinated convalescent adults. All vaccinated adults had received three doses of the BNT162b2 COVID-19 vaccine at 6.2-10.9 months prior to their inclusion to the study. All the unvaccinated participants were tested negative with QFN. Regarding the vaccinated population, 50%(8/16) of the vaccinated uninfected adults and 57.1%(8/14) of the vaccinated convalescent adults were tested positive. QFN did not detect T cellular responses in unvaccinated individuals and in a significant number of vaccinated individuals. Further comparative studies with different immunoassays are required to elucidate whether this is the result of waning immunity or low sensitivity of the assay.
Collapse
|
12
|
Kanokudom S, Chansaenroj J, Suntronwong N, Assawakosri S, Yorsaeng R, Nilyanimit P, Aeemjinda R, Khanarat N, Vichaiwattana P, Klinfueng S, Thongmee T, Katanyutanon A, Thanasopon W, Arayapong J, Withaksabut W, Srimuan D, Thatsanatorn T, Sudhinaraset N, Wanlapakorn N, Honsawek S, Poovorawan Y. Safety and immunogenicity of a third dose of COVID-19 protein subunit vaccine (Covovax TM) after homologous and heterologous two-dose regimens. Int J Infect Dis 2023; 126:64-72. [PMID: 36427701 PMCID: PMC9678824 DOI: 10.1016/j.ijid.2022.11.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVES To report the safety and immunogenicity profile of a protein subunit vaccine (CovovaxTM) given as a third (booster) dose to individuals primed with different primary vaccine regimens. METHODS A third dose was administered to individuals with an interval range of 3-10 months after the second dose. The four groups were classified according to their primary vaccine regimens, including two-dose BBIBP-CorV, AZD1222, BNT162b2, and CoronaVac/AZD1222. Immunogenicity analysis was performed to determine binding antibodies, neutralizing activity, and the T-cell responses. RESULTS Overall, 210 individuals were enrolled and boosted with the CovovaxTM vaccine. The reactogenicity was mild to moderate. Most participants elicited a high level of binding and neutralizing antibody against Wild-type and Omicron variants after the booster dose. In participants who were antinucleocapsid immunoglobulin G-negative from all groups, a booster dose could elicit neutralizing activity to Wild-type and Omicron variants by more than 95% and 70% inhibition at 28 days, respectively. The CovovaxTM vaccine could elicit a cell-mediated immune response. CONCLUSION The protein subunit vaccine (CovovaxTM) can be proposed as a booster dose after two different priming dose regimens. It has strong immunogenicity and good safety profiles.
Collapse
Affiliation(s)
- Sitthichai Kanokudom
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Center of Excellence in Osteoarthritis and Musculoskeleton, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Jira Chansaenroj
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nungruthai Suntronwong
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Suvichada Assawakosri
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Center of Excellence in Osteoarthritis and Musculoskeleton, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Ritthideach Yorsaeng
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Pornjarim Nilyanimit
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Ratchadawan Aeemjinda
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nongkanok Khanarat
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Preeyaporn Vichaiwattana
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Sirapa Klinfueng
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Thanunrat Thongmee
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | | | | | | | - Donchida Srimuan
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Thaksaporn Thatsanatorn
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Natthinee Sudhinaraset
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nasamon Wanlapakorn
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Sittisak Honsawek
- Center of Excellence in Osteoarthritis and Musculoskeleton, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand.
| | - Yong Poovorawan
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Fellow of Royal Society of Thailand (FRS[T]), the Royal Society of Thailand, Sanam Sueapa, Dusit, Bangkok, Thailand.
| |
Collapse
|
13
|
Mihaylova A, Lesichkova S, Baleva M, Nikolova‐Vlahova M, Kundurzhiev T, Kolevski A, Naumova E. Durability of humoral and cell-mediated immune response after SARS-CoV-2 mRNA vaccine administration. J Med Virol 2023; 95:e28360. [PMID: 36448089 PMCID: PMC9878094 DOI: 10.1002/jmv.28360] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/17/2022] [Accepted: 11/18/2022] [Indexed: 12/05/2022]
Abstract
Vaccination against the SARS-Cov-2 virus is an effective way to protect against the disease and the severe course of COVID-19. Forty-nine fully vaccinated with mRNA vaccines (BNT162b2 or mRNA-1273) SARS-CoV-2 infection-naïve volunteers aged 33-89 were enrolled in the study. Evaluation of the cellular and humoral immune response was performed within 1 to 3 months (T1) and 6-9 months (T2) after the second injection, and within 2-3 months (T3) after a booster dose. Additionally, a comparative analysis of the specific immune status was made between two age groups-below 60 (n = 22) and over 60 (n = 27) years. SARS-CoV-2-specific T-cell response was evaluated by IFN-γ-producing spot forming cells (SFCs) using a standardized ELISPOT assay. Virus neutralizing antibodies (VNA) against SARS-CoV-2 were measured by a blocking ELISA test and spike protein specific IgG (S-IgG) and IgA (S-IgA) antibodies-by semiquantitative ELISA. IFN-γ-producing SFCs, S-IgG, S-IgA and VNA significantly decreased 6-9 months after the second dose. After the third injection S-IgG and S-IgA markedly increased compared to T2 and reached the levels at T1. Of note, the highest values of VNA were observed at T3. No differences in the tested immune parameters were found between the two age groups. Data obtained showed that for a long period-6-9 months after a full course of immunization with mRNA vaccine, immune reactivity is present, but both cellular and humoral immune responses gradually decrease. The administration of a third dose mainly restores the specific humoral immune response against the SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Anastasiya Mihaylova
- Clinic of Clinical Immunology and Stem Cell BankUniversity Hospital AlexandrovskaSofiaBulgaria
| | - Spaska Lesichkova
- Clinic of Clinical Immunology and Stem Cell BankUniversity Hospital AlexandrovskaSofiaBulgaria
- Department of Clinical ImmunologyMedical UniversitySofiaBulgaria
| | | | - Milena Nikolova‐Vlahova
- Clinic of NephrologyUniversity Hospital St.Ivan RilskiSofiaBulgaria
- Department of Internal MedicineMedical UniversitySofiaBulgaria
| | - Todor Kundurzhiev
- Department of Occupational Medicine, Biostatistics and Medical InformaticsMedical UniversitySofiaBulgaria
| | - Alexander Kolevski
- Laboratory of MicrobiologyUniversity Hospital AlexandrovskaSofiaBulgaria
| | - Elissaveta Naumova
- Clinic of Clinical Immunology and Stem Cell BankUniversity Hospital AlexandrovskaSofiaBulgaria
- Department of Clinical ImmunologyMedical UniversitySofiaBulgaria
| |
Collapse
|
14
|
Dynamics of Antibody and T Cell Immunity against SARS-CoV-2 Variants of Concern and the Impact of Booster Vaccinations in Previously Infected and Infection-Naïve Individuals. Vaccines (Basel) 2022; 10:vaccines10122132. [PMID: 36560542 PMCID: PMC9784197 DOI: 10.3390/vaccines10122132] [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: 11/17/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Despite previous coronavirus disease 2019 (COVID-19) vaccinations and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections, SARS-CoV-2 still causes a substantial number of infections due to the waning of immunity and the emergence of new variants. Here, we assessed the SARS-CoV-2 spike subunit 1 (S1)-specific T cell responses, anti-SARS-CoV-2 receptor-binding domain (RBD) IgG serum concentrations, and the neutralizing activity of serum antibodies before and one, four, and seven months after the BNT162b2 or mRNA-1273 booster vaccination in a cohort of previously infected and infection-naïve healthcare workers (HCWs). Additionally, we assessed T cell responses against the spike protein of the SARS-CoV-2 Delta, Omicron BA.1 and BA.2 variants of concern (VOC). We found that S1-specific T cell responses, anti-RBD IgG concentrations, and neutralizing activity significantly increased one month after booster vaccination. Four months after booster vaccination, T cell and antibody responses significantly decreased but levels remained steady thereafter until seven months after booster vaccination. After a similar number of vaccinations, previously infected individuals had significantly higher S1-specific T cell, anti-RBD IgG, and neutralizing IgG responses than infection-naïve HCWs. Strikingly, we observed overall cross-reactive T cell responses against different SARS-CoV-2 VOC in both previously infected and infection-naïve HCWs. In summary, COVID-19 booster vaccinations induce strong T cell and neutralizing antibody responses and the presence of T cell responses against SARS-CoV-2 VOC suggest that vaccine-induced T cell immunity offers cross-reactive protection against different VOC.
Collapse
|
15
|
Busà R, Miele M, Sorrentino MC, Amico G, Timoneri F, Miceli V, Di Bella M, Russelli G, Gallo A, Zito G, Iannolo G, Conaldi PG, Bulati M. Long-Term Effectiveness of BNT162b2 Pfizer-BioNTech mRNA-Based Vaccine on B Cell Compartment: Efficient Recall of SARS-CoV-2-Specific Memory B Cells. Int J Mol Sci 2022; 23:15046. [PMID: 36499373 PMCID: PMC9738945 DOI: 10.3390/ijms232315046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/04/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
At present, there is a lack of clinical evidence about the impact and long-term durability of the immune response induced by the third dose of mRNA vaccines. In this study, we followed up the B cell compartment behavior in a cohort of immunocompetent individuals three and six months after the third dose of vaccine. During this period, some subjects contracted the virus. In uninfected vaccinated subjects, we did not report any changes in serum spike-specific IgG levels, with a significant reduction in IgA. Instead, subjects recovered from natural infection showed a significant increase in both specific IgG and IgA. Moreover, we showed a time-related decrease in IgG neutralizing potential to all SARS-CoV-2 variants of concern (VOC) in uninfected compared to recovered subjects, who displayed an increased neutralizing ability, particularly against the omicron variant. Finally, we underlined the presence of a pool of SARS-CoV-2-specific B cells in both groups that are prone to respond to restimulation, as demonstrated by their ability to differentiate into plasma cells and to produce anti-SARS-CoV-2-specific immunoglobulins. These data lead us to assert the long-term effectiveness of the BNT162b2 vaccine in contrasting the severe form of the pathology and prevent COVID-19-associated hospitalization.
Collapse
Affiliation(s)
- Rosalia Busà
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy
| | - Monica Miele
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy
- Ri.MED Foundation, 90133 Palermo, Italy
| | - Maria Concetta Sorrentino
- Department of Laboratory Medicine and Advanced Biotechnologies, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy
| | - Giandomenico Amico
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy
- Ri.MED Foundation, 90133 Palermo, Italy
| | - Francesca Timoneri
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy
- Ri.MED Foundation, 90133 Palermo, Italy
| | - Vitale Miceli
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy
| | - Mariangela Di Bella
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy
- Ri.MED Foundation, 90133 Palermo, Italy
| | - Giovanna Russelli
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy
| | - Alessia Gallo
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy
| | - Giovanni Zito
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy
| | - Gioacchin Iannolo
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy
| | - Pier Giulio Conaldi
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy
| | - Matteo Bulati
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy
| |
Collapse
|
16
|
SARS-CoV-2 Specific Humoral Immune Responses after BNT162b2 Vaccination in Hospital Healthcare Workers. Vaccines (Basel) 2022; 10:vaccines10122038. [PMID: 36560450 PMCID: PMC9782529 DOI: 10.3390/vaccines10122038] [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: 10/20/2022] [Revised: 11/16/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND COVID-19 pandemic has led to a loss of human life in millions and devastating socio-economic consequences worldwide. So far, vaccination is the most effective long-term strategy to control and prevent severe COVID-19 disease. The aim of the current study was to evaluate the humoral immune responses raised against the BNT162b2 vaccine in hospital healthcare workers. METHODS Total number of 173 healthcare workers enrolled in the study. Their blood samples were collected in three different time intervals after the second SARS-CoV-2 vaccination and evaluated by the ELISA method to detect anti-spike protein IgM and IgG antibodies. The baseline characteristics of all participants were collected using questionnaires and were evaluated for finding any significant data. RESULTS Our results demonstrated that the levels of antibodies were higher in the young group (21-30 years old) and also among male participants. Moreover, the highest levels of antibodies were detected from the group that received the third shot vaccination. CONCLUSIONS Our results indicate that age, gender and third-dose vaccination can affect the levels of humoral immune responses against the BNT162b2 vaccine in healthcare workers.
Collapse
|
17
|
Host Protective Immunity against Severe Acute Respiratory Coronavirus 2 (SARS-CoV-2) and the COVID-19 Vaccine-Induced Immunity against SARS-CoV-2 and Its Variants. Viruses 2022; 14:v14112541. [PMID: 36423150 PMCID: PMC9697230 DOI: 10.3390/v14112541] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
The world is now apparently at the last/recovery stage of the COVID-19 pandemic, starting from 29 December 2019, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). With the progression of time, several mutations have taken place in the original SARS-CoV-2 Wuhan strain, which have generated variants of concern (VOC). Therefore, combatting COVID-19 has required the development of COVID-19 vaccines using several platforms. The immunity induced by those vaccines is vital to study in order to assure total protection against SARS-CoV-2 and its emerging variants. Indeed, understanding and identifying COVID-19 protection mechanisms or the host immune responses are of significance in terms of designing both new and repurposed drugs as well as the development of novel vaccines with few to no side effects. Detecting the immune mechanisms for host protection against SARS-CoV-2 and its variants is crucial for the development of novel COVID-19 vaccines as well as to monitor the effectiveness of the currently used vaccines worldwide. Immune memory in terms of the production of neutralizing antibodies (NAbs) during reinfection is also very crucial to formulate the vaccine administration schedule/vaccine doses. The response of antigen-specific antibodies and NAbs as well as T cell responses, along with the protective cytokine production and the innate immunity generated upon COVID-19 vaccination, are discussed in the current review in comparison to the features of naturally induced protective immunity.
Collapse
|
18
|
Özbay Kurt FG, Lepper A, Gerhards C, Roemer M, Lasser S, Arkhypov I, Bitsch R, Bugert P, Altevogt P, Gouttefangeas C, Neumaier M, Utikal J, Umansky V. Booster dose of mRNA vaccine augments waning T cell and antibody responses against SARS-CoV-2. Front Immunol 2022; 13:1012526. [PMID: 36311732 PMCID: PMC9597683 DOI: 10.3389/fimmu.2022.1012526] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/26/2022] [Indexed: 12/03/2022] Open
Abstract
A gradual decay in humoral and cellular immune responses over time upon SAR1S-CoV-2 vaccination may cause a lack of protective immunity. We conducted a longitudinal analysis of antibodies, T cells, and monocytes in 25 participants vaccinated with mRNA or ChAdOx1-S up to 12 weeks after the 3rd (booster) dose with mRNA vaccine. We observed a substantial increase in antibodies and CD8 T cells specific for the spike protein of SARS-CoV-2 after vaccination. Moreover, vaccination induced activated T cells expressing CD69, CD137 and producing IFN-γ and TNF-α. Virus-specific CD8 T cells showed predominantly memory phenotype. Although the level of antibodies and frequency of virus-specific T cells reduced 4-6 months after the 2nd dose, they were augmented after the 3rd dose followed by a decrease later. Importantly, T cells generated after the 3rd vaccination were also reactive against Omicron variant, indicated by a similar level of IFN-γ production after stimulation with Omicron peptides. Breakthrough infection in participants vaccinated with two doses induced more SARS-CoV-2-specific T cells than the booster vaccination. We found an upregulation of PD-L1 expression on monocytes but no accumulation of myeloid cells with MDSC-like immunosuppressive phenotype after the vaccination. Our results indicate that the 3rd vaccination fosters antibody and T cell immune response independently from vaccine type used for the first two injections. However, such immune response is attenuated over time, suggesting thereby the need for further vaccinations.
Collapse
Affiliation(s)
- Feyza Gül Özbay Kurt
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Alisa Lepper
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Catharina Gerhards
- Institute for Clinical Chemistry, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Mathis Roemer
- Institute for Clinical Chemistry, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Samantha Lasser
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Ihor Arkhypov
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Rebekka Bitsch
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Peter Bugert
- German Red Cross Blood Service Baden-Württemberg – Hessen, Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Peter Altevogt
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Cécile Gouttefangeas
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Michael Neumaier
- Institute for Clinical Chemistry, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
19
|
Tani N, Ikematsu H, Goto T, Gondo K, Inoue T, Yanagihara Y, Kurata Y, Oishi R, Minami J, Onozawa K, Nagano S, Kuwano H, Akashi K, Shimono N, Chong Y. Correlation of Postvaccination Fever With Specific Antibody Response to Severe Acute Respiratory Syndrome Coronavirus 2 BNT162b2 Booster and No Significant Influence of Antipyretic Medication. Open Forum Infect Dis 2022; 9:ofac493. [PMID: 36267253 PMCID: PMC9578158 DOI: 10.1093/ofid/ofac493] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/22/2022] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND A severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mRNA vaccine booster elicits sufficient antibody responses that protect against coronavirus disease 2019, whereas adverse reactions such as fever have been commonly reported. Associations between adverse reactions and antibody responses have not been fully characterized, nor has the influence of antipyretic use. METHODS This is a prospective observational cohort study in Japan, following our prior investigation of BNT162b2 2-dose primary series. Spike-specific immunoglobulin G (IgG) titers were measured for SARS-CoV-2-naive hospital healthcare workers who received a BNT162b2 booster. The severity of solicited adverse reactions, including the highest body temperature, and self-medicated antipyretics were reported daily for 7 days following vaccination through a web-based self-reporting diary. RESULTS The data of 281 healthcare workers were available. Multivariate analysis extracted fever after the booster dose (β = .305, P < .001) as being significantly correlated with the specific IgG titers. The analysis of 164 participants with data from the primary series showed that fever after the second dose was associated with the emergence of fever after the booster dose (relative risk, 3.97 [95% confidence interval, 2.48-6.35]); however, the IgG titers after the booster dose were not associated with the presence or degree of fever after the second dose. There were no significant differences in the IgG titers by the use, type, or dosage of antipyretic medication. CONCLUSIONS These results suggest an independent correlation between mRNA vaccine-induced specific IgG levels and post-booster vaccination fever, without any significant influence of fever after the primary series. Antipyretic medications for adverse reactions should not interfere with the elevation of specific IgG titers.
Collapse
Affiliation(s)
- Naoki Tani
- Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | | | - Takeyuki Goto
- Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kei Gondo
- Clinical Laboratory, Fukuoka City Hospital, Fukuoka, Japan
| | - Takeru Inoue
- Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | | | | | - Ryo Oishi
- Department of Infectious Diseases, Fukuoka City Hospital, Fukuoka, Japan
| | - Junya Minami
- Department of Infectious Diseases, Fukuoka City Hospital, Fukuoka, Japan
| | - Kyoko Onozawa
- Department of Infectious Diseases, Fukuoka City Hospital, Fukuoka, Japan
| | - Sukehisa Nagano
- Department of Neurology, Fukuoka City Hospital, Fukuoka, Japan
| | - Hiroyuki Kuwano
- Department of Surgery, Fukuoka City Hospital, Fukuoka, Japan
| | - Koichi Akashi
- Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Nobuyuki Shimono
- Center for the Study of Global Infection, Kyushu University Hospital, Fukuoka, Japan
| | - Yong Chong
- Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| |
Collapse
|
20
|
Franzese M, Coppola L, Silva R, Santini SA, Cinquanta L, Ottomano C, Salvatore M, Incoronato M. SARS-CoV-2 antibody responses before and after a third dose of the BNT162b2 vaccine in Italian healthcare workers aged ≤60 years: One year of surveillance. Front Immunol 2022; 13:947187. [PMID: 36248864 PMCID: PMC9566572 DOI: 10.3389/fimmu.2022.947187] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 09/02/2022] [Indexed: 11/29/2022] Open
Abstract
This study monitored the anti-spike-receptor-binding domain (RBD) and neutralizing antibodies induced by the Pfizer/BioNTech mRNA BNT162b2 vaccine in a cohort of 163 healthcare workers aged ≤60 years. We have taken advantage of two study groups, both of whom received the first two doses in the same time window, but Group 1 (54 HCWs) received the third dose 2 months before Group 2 (68 HCWs) did. The cohorts were monitored from the 12th day after the first vaccine dose up to 1 month after the third vaccine dose for a total of eight time points and about 1 year of surveillance (T1 = 12 days after the first dose; T2 = 10 days after the second dose; T3 = 1 month after the second dose; T4 = 3 months after the second dose; T5 = 4 months after the second dose; T6 = 5 months after the second dose; T7 = 7 months after the second dose; T8 = 1 month after the third dose for Group 1; T8* = 9 months after the second dose for Group 2; T9 = 1 month after the third dose for Group 2). The mean value of anti-spike antibodies decreased faster over time, but at T7, its decline was significantly slowed (T7 vs. T8*). After the third dose, the anti-spike titer rose about 34-fold (T7 vs. T8 and T8* vs. T9) and the booster improved the anti-spike titer by about three times compared with that of the second dose (T3 vs. T8 and T3 vs. T9), and no difference was noted between the two groups. The neutralizing titer was evaluated at T3, T7, T8, and T9. Anti-spike and neutralizing antibodies were found to be strongly correlated (r2 = 0.980; p < 0.001). At T3, 70% of the participants had a neutralizing antibody titer >91% of total anti-spike antibodies that increased to 90% after the third dose (T8 and T9). However, when the anti-spike titer reached its lowest value (T7), the neutralizing antibody levels decreased even further, representing only 44% of total anti-spike antibodies (p < 0.0001). Our findings show that the third vaccine dose improves the humoral response, but the wane of the anti-spike and neutralizing antibody titers over time is more marked in the neutralizing antibodies.
Collapse
Affiliation(s)
- Monica Franzese
- Research Laboratory, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Synlab SDN Spa, Naples, Italy
| | - Luigi Coppola
- Research Laboratory, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Synlab SDN Spa, Naples, Italy
| | - Romina Silva
- Research Laboratory, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Synlab SDN Spa, Naples, Italy
| | | | - Luigi Cinquanta
- Research Laboratory, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Synlab SDN Spa, Naples, Italy
| | - Cosimo Ottomano
- Research Laboratory, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Synlab SDN Spa, Naples, Italy
| | - Marco Salvatore
- Research Laboratory, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Synlab SDN Spa, Naples, Italy
| | - Mariarosaria Incoronato
- Research Laboratory, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Synlab SDN Spa, Naples, Italy
- *Correspondence: Mariarosaria Incoronato,
| |
Collapse
|
21
|
Rzymski P, Kasianchuk N, Sikora D, Poniedziałek B. COVID-19 vaccinations and rates of infections, hospitalizations, ICU admissions, and deaths in Europe during SARS-CoV-2 Omicron wave in the first quarter of 2022. J Med Virol 2022; 95:e28131. [PMID: 36068643 PMCID: PMC9537885 DOI: 10.1002/jmv.28131] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/11/2022] [Accepted: 09/04/2022] [Indexed: 01/11/2023]
Abstract
The vaccination campaigns brought hope to minimizing the coronavirus disease 2019 (COVID-19) burden. However, the emergence of novel, highly transmissible Omicron lineage of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the waning of neutralizing antibodies a few months after vaccination has brought concerns over the vaccine efficacy. The present work analyzed the relationships between COVID-19 vaccine coverage (completion of primary course and booster dose intake) in the European Economic Area and rates of infection, hospitalizations, admissions to intensive care units (ICU), and deaths during the Omicron wave in the first quarter of 2022 (January-April). As demonstrated, infection rates were not correlated to vaccine coverage in any considered month. For January and February, the rates of hospitalizations, intensive care unit (ICU) admissions, and death due to COVID-19 were strongly negatively correlated (r =- 0.54 to -0.82) with the percentage of individuals who completed initial vaccination protocol and the percentage of those who received a booster dose. However, in March and April, the percentage of the population with primary vaccination course correlated negatively only with ICU admissions (r = -0.77 and -0.46, respectively). The uptake of boosters in March still remained in significant negative correlation with hospitalizations (r = -0.45), ICU admissions (r = -0.70) and deaths due to COVID-19 (r = -0.37), although in April these relationships were no longer observed. The percentage of individuals with confirmed SARS-CoV-2 infection did not correlate with the pandemic indices for any considered month. The study indicates that COVID-19 vaccination, including booster administration, was beneficial in decreasing the overwhelming of healthcare systems during the Omicron wave, but novel vaccine strategies may be required in the long term to enhance the effectiveness and durability of vaccine-induced protection during future waves of SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Piotr Rzymski
- Department of Environmental MedicinePoznan University of Medical SciencesPoznańPoland,Integrated Science Association (ISA)Universal Scientific Education and Research Network (USERN)PoznańPoland
| | - Nadiia Kasianchuk
- Faculty of BiologyAdam Mickiewicz UniversityPoznańPoland,Faculty of PharmacyBogomolets National Medical UniversityKyivUkraine
| | - Dominika Sikora
- Department of Environmental MedicinePoznan University of Medical SciencesPoznańPoland,Doctoral SchoolPoznan University of Medical SciencesPoznańPoland
| | - Barbara Poniedziałek
- Department of Environmental MedicinePoznan University of Medical SciencesPoznańPoland
| |
Collapse
|
22
|
Saresella M, Piancone F, Marventano I, Hernis A, Trabattoni D, Invernizzi M, La Rosa F, Clerici M. Innate immune responses to three doses of the BNT162b2 mRNA SARS-CoV-2 vaccine. Front Immunol 2022; 13:947320. [PMID: 36072604 PMCID: PMC9443429 DOI: 10.3389/fimmu.2022.947320] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/01/2022] [Indexed: 11/23/2022] Open
Abstract
To explore the effects of SARS-CoV-2-mRNA vaccines on innate immune responses we enrolled 58 individuals who received 3 doses of the BNT162b2 vaccine in a longitudinal study; 45 of these individuals had never been SARS-CoV-2 infected. Results showed that vaccination significantly increased: 1) classical and intermediate inflammatory monocytes, 2) CD56bright, CD56dim, and CD56dim/CD16dim NK cells, and 3) IFN-γ+ ;production as well as perforin and granzyme content by NK cells. Vaccination also reduced expression of the NK inhibitory receptor ILT-2, increasing that of the stimulatory molecule 2DS2. These effects were long-lasting and were boosted by every vaccine dose. Notably, ILT-2 expressing NK cells were reduced even more robustly in COVID-19-recovereed vaccines. BNT162b1 mRNA vaccine is known to induce potent adaptive immune responses; results herein show its ability to modulate innate immune responses as well, offering further support to the indication to proceed with worldwide vaccination efforts to end the SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Marina Saresella
- IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
- *Correspondence: Marina Saresella,
| | | | | | - Ambra Hernis
- IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
| | - Daria Trabattoni
- Departments of Biomedical and Clinical Sciences L. Sacco, University of Milan, Milan, Italy
| | | | | | - Mario Clerici
- IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
- Pathophysiology and Transplantation, University of Milan, Milan, Italy
| |
Collapse
|
23
|
Almendro-Vázquez P, Chivite-Lacaba M, Utrero-Rico A, González-Cuadrado C, Laguna-Goya R, Moreno-Batanero M, Sánchez-Paz L, Luczkowiak J, Labiod N, Folgueira MD, Delgado R, Paz-Artal E. Cellular and humoral immune responses and breakthrough infections after three SARS-CoV-2 mRNA vaccine doses. Front Immunol 2022; 13:981350. [PMID: 36059485 PMCID: PMC9428395 DOI: 10.3389/fimmu.2022.981350] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Background SARS-CoV-2 vaccination has proven the most effective measure to control the COVID-19 pandemic. Booster doses are being administered with limited knowledge on their need and effect on immunity. Objective To determine the duration of specific T cells, antibodies and neutralization after 2-dose vaccination, to assess the effect of a third dose on adaptive immunity and to explore correlates of protection against breakthrough infection. Methods 12-month longitudinal assessment of SARS-CoV-2-specific T cells, IgG and neutralizing antibodies triggered by 2 BNT162b2 doses followed by a third mRNA-1273 dose in a cohort of 77 healthcare workers: 17 with SARS-CoV-2 infection prior to vaccination (recovered) and 60 naïve. Results Peak levels of cellular and humoral response were achieved 2 weeks after the second dose. Antibodies declined thereafter while T cells reached a plateau 3 months after vaccination. The decline in neutralization was specially marked in naïve individuals and it was this group who benefited most from the third dose, which resulted in a 20.9-fold increase in neutralization. Overall, recovered individuals maintained higher levels of T cells, antibodies and neutralization 1 to 6 months post-vaccination than naïve. Seventeen asymptomatic or mild SARS-CoV-2 breakthrough infections were reported during follow-up, only in naïve individuals. This viral exposure boosted adaptive immunity. High peak levels of T cells and neutralizing antibodies 15 days post-vaccination associated with protection from breakthrough infections. Conclusion Booster vaccination in naïve individuals and the inclusion of viral antigens other than spike in future vaccine formulations could be useful strategies to prevent SARS-CoV-2 breakthrough infections.
Collapse
Affiliation(s)
- Patricia Almendro-Vázquez
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- *Correspondence: Patricia Almendro-Vázquez,
| | - Marta Chivite-Lacaba
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Alberto Utrero-Rico
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | | | - Rocio Laguna-Goya
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas (CIBERINFEC – Instituto de Salud Carlos III), Madrid, Spain
| | | | - Laura Sánchez-Paz
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Joanna Luczkowiak
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Nuria Labiod
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - María Dolores Folgueira
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Department of Microbiology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Rafael Delgado
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas (CIBERINFEC – Instituto de Salud Carlos III), Madrid, Spain
- Department of Microbiology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Department of Medicine, Medical School, Universidad Complutense de Madrid, Madrid, Spain
| | - Estela Paz-Artal
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas (CIBERINFEC – Instituto de Salud Carlos III), Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Medical School, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
24
|
Miele M, Busà R, Russelli G, Sorrentino MC, Di Bella M, Timoneri F, Vitale G, Calzolari E, Vitulo P, Mularoni A, Conaldi PG, Bulati M. Analysis of the Specific Immune Response after the Third Dose of mRNA COVID-19 Vaccines in Organ Transplant Recipients: Possible Spike-S1 Reactive IgA Signature in Protection from SARS-CoV-2 Infection. Microorganisms 2022; 10:microorganisms10081563. [PMID: 36013981 PMCID: PMC9415050 DOI: 10.3390/microorganisms10081563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/25/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Several studies have indicated that anti-SARS-CoV-2 mRNA vaccinations are less effective in inducing robust immune responses among solid organ transplant recipients (SOTRs) compared with the immunocompetent. The third dose of vaccine in SOTRs showed promising results of immunogenicity, even though clinical studies have suggested that immunocompromised subjects are less likely to build a protective immune response against SARS-CoV-2 resulting in lower vaccine efficacy for the prevention of severe COVID-19. Methods: Serological IgG and IgA were analyzed through CLIA or ELISA, respectively, while Spike-specific T cells were detected by ELISpot assay after the second and third dose of vaccine in 43 SOTRs. Results: The third dose induced an improvement in antibody response against SARS-CoV-2. We also reported a strong correlation between specific humoral and cellular responses after the third dose, even though we did not see significant changes in the magnitude of the SARS-CoV-2-specific T cell response. SOTRs who contracted the SARS-CoV-2 infection after the third dose, despite eliciting a positive IgG response, failed to mount an anti-Spike-S1 IgA response, both after the third dose and after SARS-CoV-2 infection. Conclusions: We can conclude that serum IgA detection can be helpful, along with IgG detection, for the evaluation of vaccine efficacy, principally in fragile subjects at high risk of infection.
Collapse
Affiliation(s)
- Monica Miele
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy; (R.B.); (G.R.); (M.D.B.); (F.T.); (G.V.); (P.G.C.); (M.B.)
- Ri.MED Foundation, 90133 Palermo, Italy
- Correspondence:
| | - Rosalia Busà
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy; (R.B.); (G.R.); (M.D.B.); (F.T.); (G.V.); (P.G.C.); (M.B.)
| | - Giovanna Russelli
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy; (R.B.); (G.R.); (M.D.B.); (F.T.); (G.V.); (P.G.C.); (M.B.)
| | - Maria Concetta Sorrentino
- Department of Laboratory Medicine and Advanced Biotechnologies, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy;
| | - Mariangela Di Bella
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy; (R.B.); (G.R.); (M.D.B.); (F.T.); (G.V.); (P.G.C.); (M.B.)
- Ri.MED Foundation, 90133 Palermo, Italy
| | - Francesca Timoneri
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy; (R.B.); (G.R.); (M.D.B.); (F.T.); (G.V.); (P.G.C.); (M.B.)
- Ri.MED Foundation, 90133 Palermo, Italy
| | - Giampiero Vitale
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy; (R.B.); (G.R.); (M.D.B.); (F.T.); (G.V.); (P.G.C.); (M.B.)
- Ri.MED Foundation, 90133 Palermo, Italy
| | - Elisa Calzolari
- Allergy and Respiratory Diseases, Department of Internal Medicine (DIMI), IRCCS Policlinico San Martino, University of Genoa, 16124 Genoa, Italy;
| | - Patrizio Vitulo
- Department for the Treatment and Study of Cardiothoracic Diseases and Cardiothoracic Transplantation, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy;
| | - Alessandra Mularoni
- Department of Infectious Diseases, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy;
| | - Pier Giulio Conaldi
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy; (R.B.); (G.R.); (M.D.B.); (F.T.); (G.V.); (P.G.C.); (M.B.)
| | - Matteo Bulati
- Research Department, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT), 90127 Palermo, Italy; (R.B.); (G.R.); (M.D.B.); (F.T.); (G.V.); (P.G.C.); (M.B.)
| |
Collapse
|
25
|
Zarębska-Michaluk D, Hu C, Brzdęk M, Flisiak R, Rzymski P. COVID-19 Vaccine Booster Strategies for Omicron SARS-CoV-2 Variant: Effectiveness and Future Prospects. Vaccines (Basel) 2022; 10:vaccines10081223. [PMID: 36016111 PMCID: PMC9412973 DOI: 10.3390/vaccines10081223] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 12/05/2022] Open
Abstract
In the light of the lack of authorized COVID-19 vaccines adapted to the Omicron variant lineage, the administration of the first and second booster dose is recommended. It remains important to monitor the efficacy of such an approach in order to inform future preventive strategies. The present paper summarizes the research progress on the effectiveness of the first and second booster doses of COVID-19. It also discusses the potential approach in vaccination strategies that could be undertaken to maintain high levels of protection during the waves of SARS-CoV-2 infections. Although this approach can be based, with some shortcomings, on the first-generation vaccines, other vaccination strategies should be explored, including developing multiple antigen-based (multivariant-adapted) booster doses with enhanced durability of immune protection, e.g., through optimization of the half-life of generated antibodies.
Collapse
Affiliation(s)
- Dorota Zarębska-Michaluk
- Department of Infectious Diseases, Jan Kochanowski University, 25-369 Kielce, Poland; (D.Z.-M.); (M.B.)
| | - Chenlin Hu
- College of Pharmacy, University of Houston, Houston, TX 77204, USA;
| | - Michał Brzdęk
- Department of Infectious Diseases, Jan Kochanowski University, 25-369 Kielce, Poland; (D.Z.-M.); (M.B.)
| | - Robert Flisiak
- Department of Infectious Diseases and Hepatology, Medical University of Białystok, 15-540 Białystok, Poland;
| | - Piotr Rzymski
- Department of Environmental Medicine, Poznan University of Medical Sciences, 60-806 Poznan, Poland
- Integrated Science Association (ISA), Universal Scientific Education and Research Network (USERN), 60-806 Poznań, Poland
- Correspondence:
| |
Collapse
|
26
|
Hassan R, Mohammed S. Evaluation of immunoglobulin G level among subjects vaccinated with different types of COVID-19 vaccines in the karbala population, Iraq. BIOMEDICAL AND BIOTECHNOLOGY RESEARCH JOURNAL (BBRJ) 2022. [DOI: 10.4103/bbrj.bbrj_213_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|