1
|
Shahid Z, Jain T, Dioverti V, Pennisi M, Mikkilineni L, Thiruvengadam SK, Shah NN, Dadwal S, Papanicolaou G, Hamadani M, Carpenter PA, Alfaro GM, Seo SK, Hill JA. Best Practice Considerations by The American Society of Transplant and Cellular Therapy: Infection Prevention and Management After Chimeric Antigen Receptor T Cell Therapy for Hematological Malignancies. Transplant Cell Ther 2024; 30:955-969. [PMID: 39084261 DOI: 10.1016/j.jtct.2024.07.018] [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: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is rapidly advancing, offering promising treatments for patients with hematological malignancy. However, associated infectious complications remain a significant concern because of their contribution to patient morbidity and non-relapse mortality. Recent epidemiological insights shed light on risk factors for infections after CAR T-cell therapy. However, the available evidence is predominantly retrospective, highlighting a need for further prospective studies. Institutions are challenged with managing infections after CAR T-cell therapy but variations in the approaches taken underscore the importance of standardizing infection prevention and management protocols across different healthcare settings. Therefore, the Infectious Diseases Special Interest Group of the American Society of Transplantation and Cellular Therapy assembled an expert panel to develop best practice considerations. The aim was to guide healthcare professionals in optimizing infection prevention and management for CAR T-cell therapy recipients and advocates for early consultation of Infectious Diseases during treatment planning phases given the complexities involved. By synthesizing current evidence and expert opinion these best practice considerations provide the basis for understanding infection risk after CAR T-cell therapies and propose risk-mitigating strategies in children, adolescents, and adults. Continued research and collaboration will be essential to refining and effectively implementing these recommendations.
Collapse
Affiliation(s)
- Zainab Shahid
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Tania Jain
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Veronica Dioverti
- Division of Infectious Disease, Department of Medicine, John Hopkins School of Medicine, Baltimore, Maryland
| | - Martini Pennisi
- Division of Hematology and Stem Cell Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Lekha Mikkilineni
- Division of Bone and Marrow Transplant & Cellular Therapies, Stanford School of Medicine, Palo Alto, California
| | - Swetha Kambhampati Thiruvengadam
- Division of Lymphoma, Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sanjeet Dadwal
- Division of Infectious Disease, Department of Medicine, City of Hope National Medical Center, Duarte, California
| | - Genovefa Papanicolaou
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mehdi Hamadani
- Bone Marrow Transplant & Cellular Therapy Program, Medical College of Wisconsin, Milwaukee, Wisconsin; Center for International Blood and Marrow Transplant Research, Milwaukee, Wisconsin
| | - Paul A Carpenter
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Gabriela Maron Alfaro
- Department of Infectious Diseases, St. Jude Children's Research Hospital and Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Susan K Seo
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joshua A Hill
- Vaccine and Infectious Disease Division, Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| |
Collapse
|
2
|
Macchia I, La Sorsa V, Ciervo A, Ruspantini I, Negri D, Borghi M, De Angelis ML, Luciani F, Martina A, Taglieri S, Durastanti V, Altavista MC, Urbani F, Mancini F. T Cell Peptide Prediction, Immune Response, and Host-Pathogen Relationship in Vaccinated and Recovered from Mild COVID-19 Subjects. Biomolecules 2024; 14:1217. [PMID: 39456150 PMCID: PMC11505848 DOI: 10.3390/biom14101217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024] Open
Abstract
COVID-19 remains a significant threat, particularly to vulnerable populations. The emergence of new variants necessitates the development of treatments and vaccines that induce both humoral and cellular immunity. This study aimed to identify potentially immunogenic SARS-CoV-2 peptides and to explore the intricate host-pathogen interactions involving peripheral immune responses, memory profiles, and various demographic, clinical, and lifestyle factors. Using in silico and experimental methods, we identified several CD8-restricted SARS-CoV-2 peptides that are either poorly studied or have previously unreported immunogenicity: fifteen from the Spike and three each from non-structural proteins Nsp1-2-3-16. A Spike peptide, LA-9, demonstrated a 57% response rate in ELISpot assays using PBMCs from 14 HLA-A*02:01 positive, vaccinated, and mild-COVID-19 recovered subjects, indicating its potential for diagnostics, research, and multi-epitope vaccine platforms. We also found that younger individuals, with fewer vaccine doses and longer intervals since infection, showed lower anti-Spike (ELISA) and anti-Wuhan neutralizing antibodies (pseudovirus assay), higher naïve T cells, and lower central memory, effector memory, and CD4hiCD8low T cells (flow cytometry) compared to older subjects. In our cohort, a higher prevalence of Vδ2-γδ and DN T cells, and fewer naïve CD8 T cells, seemed to correlate with strong cellular and lower anti-NP antibody responses and to associate with Omicron infection, absence of confusional state, and habitual sporting activity.
Collapse
Affiliation(s)
- Iole Macchia
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Valentina La Sorsa
- Research Promotion and Coordination Service, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Alessandra Ciervo
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Irene Ruspantini
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Martina Borghi
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Maria Laura De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Francesca Luciani
- National Center for the Control and Evaluation of Medicines, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.L.); (A.M.)
| | - Antonio Martina
- National Center for the Control and Evaluation of Medicines, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.L.); (A.M.)
| | - Silvia Taglieri
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Valentina Durastanti
- Neurology Unit, San Filippo Neri Hospital, ASL RM1, 00135 Rome, Italy; (V.D.); (M.C.A.)
| | | | - Francesca Urbani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Fabiola Mancini
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| |
Collapse
|
3
|
Wagstaffe HR, Thwaites RS, Reynaldi A, Sidhu JK, McKendry R, Ascough S, Papargyris L, Collins AM, Xu J, Lemm NM, Siggins MK, Chain BM, Killingley B, Kalinova M, Mann A, Catchpole A, Davenport MP, Openshaw PJM, Chiu C. Mucosal and systemic immune correlates of viral control after SARS-CoV-2 infection challenge in seronegative adults. Sci Immunol 2024; 9:eadj9285. [PMID: 38335268 DOI: 10.1126/sciimmunol.adj9285] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 01/12/2024] [Indexed: 02/12/2024]
Abstract
Human infection challenge permits in-depth, early, and pre-symptomatic characterization of the immune response, enabling the identification of factors that are important for viral clearance. Here, we performed intranasal inoculation of 34 young adult, seronegative volunteers with a pre-Alpha severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) strain. Of these participants, 18 (53%) became infected and showed an interferon-dominated mediator response with divergent kinetics between nasal and systemic sites. Peripheral CD4+ and CD8+ T cell activation and proliferation were early and robust but showed distinct kinetic and phenotypic profiles; antigen-specific T cells were largely CD38+Ki67+ and displayed central and effector memory phenotypes. Both mucosal and systemic antibodies became detectable around day 10, but nasal antibodies plateaued after day 14 while circulating antibodies continued to rise. Intensively granular measurements in nasal mucosa and blood allowed modeling of immune responses to primary SARS-CoV-2 infection that revealed CD8+ T cell responses and early mucosal IgA responses strongly associated with viral control, indicating that these mechanisms should be targeted for transmission-reducing intervention.
Collapse
Affiliation(s)
- Helen R Wagstaffe
- Department of Infectious Disease, Imperial College London, London, UK
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Jasmin K Sidhu
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Richard McKendry
- Department of Infectious Disease, Imperial College London, London, UK
| | - Stephanie Ascough
- Department of Infectious Disease, Imperial College London, London, UK
| | - Loukas Papargyris
- Department of Infectious Disease, Imperial College London, London, UK
| | - Ashley M Collins
- Department of Infectious Disease, Imperial College London, London, UK
| | - Jiayun Xu
- Department of Infectious Disease, Imperial College London, London, UK
| | - Nana-Marie Lemm
- Department of Infectious Disease, Imperial College London, London, UK
| | - Matthew K Siggins
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Benny M Chain
- UCL Division of Infection and Immunity, University College London, London, UK
| | - Ben Killingley
- Department of Infectious Diseases, University College London Hospital, London, UK
| | | | | | | | - Miles P Davenport
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | | | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, UK
| |
Collapse
|
4
|
Díaz-Dinamarca DA, Cárdenas-Cáceres S, Muena NA, Díaz P, Barra G, Puentes R, Escobar DF, Díaz-Samirin M, Santis-Alay NT, Canales C, Díaz J, García-Escorza HE, Grifoni A, Sette A, Tischler ND, Vasquez AE. Booster Vaccination with BNT162b2 Improves Cellular and Humoral Immune Response in the Pediatric Population Immunized with CoronaVac. Vaccines (Basel) 2024; 12:919. [PMID: 39204043 PMCID: PMC11359105 DOI: 10.3390/vaccines12080919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 09/03/2024] Open
Abstract
The SARS-CoV-2 Omicron variant and its sublineages continue to cause COVID-19-associated pediatric hospitalizations, severe disease, and death globally. BNT162b2 and CoronaVac are the main vaccines used in Chile. Much less is known about the Wuhan-Hu-1 strain-based vaccines in the pediatric population compared to adults. Given the worldwide need for booster vaccinations to stimulate the immune response against new Omicron variants of SARS-CoV-2, we characterized the humoral and cellular immune response against Omicron variant BA.1 in a pediatric cohort aged 10 to 16 years who received heterologous vaccination based on two doses of CoronaVac, two doses of CoronaVac (2x) plus one booster dose of BNT162b2 [CoronaVac(2x) + BNT162b2 (1x)], two doses of CoronaVac plus two booster doses of BNT162b2 [CoronaVac(2x) + BNT162b2 (2x)], and three doses of BNT162b2. We observed that the [CoronaVac(2x) + BNT162b2 (2x)] vaccination showed higher anti-S1 and neutralizing antibody titers and CD4 and CD8 T cell immunity specific to the Omicron variant compared to immunization with two doses of CoronaVac alone. Furthermore, from all groups tested, immunity against Omicron was highest in individuals who received three doses of BNT162b2. We conclude that booster vaccination with BNT162b2, compared to two doses of CoronaVac alone, induces a greater protective immunity.
Collapse
Affiliation(s)
- Diego A. Díaz-Dinamarca
- Subdepartamento Innovación y Desarrollo, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (P.D.); (G.B.); (D.F.E.)
| | - Simone Cárdenas-Cáceres
- Laboratorio de Virología Molecular, Centro Ciencia & Vida, Fundación Ciencia & Vida, Santiago 8581151, Chile; (S.C.-C.); (N.A.M.)
| | - Nicolás A. Muena
- Laboratorio de Virología Molecular, Centro Ciencia & Vida, Fundación Ciencia & Vida, Santiago 8581151, Chile; (S.C.-C.); (N.A.M.)
| | - Pablo Díaz
- Subdepartamento Innovación y Desarrollo, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (P.D.); (G.B.); (D.F.E.)
| | - Gisselle Barra
- Subdepartamento Innovación y Desarrollo, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (P.D.); (G.B.); (D.F.E.)
| | - Rodrigo Puentes
- Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7780050, Chile; (R.P.); (N.T.S.-A.); (C.C.); (J.D.)
| | - Daniel F. Escobar
- Subdepartamento Innovación y Desarrollo, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (P.D.); (G.B.); (D.F.E.)
| | - Michal Díaz-Samirin
- Subdepartamento Innovación y Desarrollo, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (P.D.); (G.B.); (D.F.E.)
| | - Natalia T. Santis-Alay
- Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7780050, Chile; (R.P.); (N.T.S.-A.); (C.C.); (J.D.)
| | - Cecilia Canales
- Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7780050, Chile; (R.P.); (N.T.S.-A.); (C.C.); (J.D.)
| | - Janepsy Díaz
- Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7780050, Chile; (R.P.); (N.T.S.-A.); (C.C.); (J.D.)
| | | | - Alba Grifoni
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; (A.G.); (A.S.)
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; (A.G.); (A.S.)
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA
| | - Nicole D. Tischler
- Laboratorio de Virología Molecular, Centro Ciencia & Vida, Fundación Ciencia & Vida, Santiago 8581151, Chile; (S.C.-C.); (N.A.M.)
- Escuela de Bioquímica, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510157, Chile
| | - Abel E. Vasquez
- Subdepartamento Innovación y Desarrollo, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (P.D.); (G.B.); (D.F.E.)
| |
Collapse
|
5
|
Eviatar T, Pappo A, Freund T, Friedlander Y, Elkayam O, Hagin D, Heshin-Bekenstein M. Cellular immune response to the anti-SARS-CoV-2 BNT162b2 mRNA vaccine in pediatric autoimmune inflammatory rheumatic disease patients and controls. Clin Exp Immunol 2024; 217:167-172. [PMID: 38767466 PMCID: PMC11239557 DOI: 10.1093/cei/uxae044] [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: 02/23/2024] [Revised: 04/03/2024] [Accepted: 05/18/2024] [Indexed: 05/22/2024] Open
Abstract
This paper aims to compare the cellular immune response to the SARS-CoV-2 BNT162b2 vaccine of pediatric patients with autoimmune inflammatory rheumatic disease (pAIIRD) and healthy controls. A prospective longitudinal study was conducted between April 2021 and December 2022 at the Tel Aviv Medical Center. Children <18 years, with pediatric-onset AIIRD and healthy controls, who have received at least two doses of the BNT162b2 vaccine, were included. Humoral response was evaluated by serum levels of anti-SARS-CoV-2 receptor-binding domain antibodies. Cellular response was evaluated by flow cytometry, measuring IFNγ and TNFα production by CD4+ T cells following stimulation with SARS-CoV-2 Spike peptide mix. The study included 20 pAIIRD patients and 11 controls. The mean age of participants was 12.6 ± 2.94 years, with 58.1% females. The cellular response to the BNT162b2 vaccine was statistically similar in both groups. However, the humoral response was statistically lower in pAIIRD compared with the healthy control group. There was no statistically significant correlation between the humoral response and cellular response. During the study period, 43.75% of AIIRD children and 72.7% of controls had a breakthrough COVID-19 infection (P = 0.48). Bivariate models examining the effect of the cellular response and presence of an AIIRD on breakthrough infections found no effect. Compared with healthy controls, pAIIRD demonstrated similar cellular responses. Patients showed reduced humoral response compared with healthy adolescents, but similar breakthrough infection rates. These findings may support the importance of the cellular response in protecting against COVID-19 infections.
Collapse
Affiliation(s)
- Tali Eviatar
- Rheumatology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Adi Pappo
- Pediatric Rheumatology Unit, Schneider Children’s Hospital, Petach Tikva, Israel
| | - Tal Freund
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Allergy and Clinical Immunology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | | | - Ori Elkayam
- Rheumatology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - David Hagin
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Allergy and Clinical Immunology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Merav Heshin-Bekenstein
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Pediatric Rheumatology Service, Dana Children’s Hospital of Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| |
Collapse
|
6
|
Mouton W, Oriol G, Compagnon C, Saade C, Saker K, Franc P, Mokdad B, Fleurie A, Lacoux X, Daniel S, Berthier F, Barnel C, Pozzetto B, Fassier JB, Dubois V, Djebali S, Dubois M, Walzer T, Marvel J, Brengel-Pesce K, Trouillet-Assant S. Combining SARS-CoV-2 interferon-gamma release assay with humoral response assessment to define immune memory profiles. Eur J Immunol 2024; 54:e2451035. [PMID: 38627984 DOI: 10.1002/eji.202451035] [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: 01/26/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 07/07/2024]
Abstract
OBJECTIVES In the post-SARS-CoV-2 pandemic era, "breakthrough infections" are still documented, due to variants of concerns (VoCs) emergence and waning humoral immunity. Despite widespread utilization, the definition of the anti-Spike (S) immunoglobulin-G (IgG) threshold to define protection has unveiled several limitations. Here, we explore the advantages of incorporating T-cell response assessment to enhance the definition of immune memory profile. METHODS SARS-CoV-2 interferon-gamma release assay test (IGRA) was performed on samples collected longitudinally from immunocompetent healthcare workers throughout their immunization by infection and/or vaccination, anti-receptor-binding domain IgG levels were assessed in parallel. The risk of symptomatic infection according to cellular/humoral immune capacities during Omicron BA.1 wave was then estimated. RESULTS Close to 40% of our samples were exclusively IGRA-positive, largely due to time elapsed since their last immunization. This suggests that individuals have sustained long-lasting cellular immunity, while they would have been classified as lacking protective immunity based solely on IgG threshold. Moreover, the Cox regression model highlighted that Omicron BA.1 circulation raises the risk of symptomatic infection while increased anti-receptor-binding domain IgG and IGRA levels tended to reduce it. CONCLUSION The discrepancy between humoral and cellular responses highlights the significance of assessing the overall adaptive immune response. This integrated approach allows the identification of vulnerable subjects and can be of interest to guide antiviral prophylaxis at an individual level.
Collapse
Affiliation(s)
- William Mouton
- CIRI - Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Lyon, France
- Joint Research Unit Hospices Civils de Lyon-bioMerieux S.A., Hôpital Lyon Sud, Pierre-Bénite, France
| | - Guy Oriol
- Joint Research Unit Hospices Civils de Lyon-bioMerieux S.A., Hôpital Lyon Sud, Pierre-Bénite, France
| | - Christelle Compagnon
- Joint Research Unit Hospices Civils de Lyon-bioMerieux S.A., Hôpital Lyon Sud, Pierre-Bénite, France
| | - Carla Saade
- CIRI - Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Lyon, France
- Joint Research Unit Hospices Civils de Lyon-bioMerieux S.A., Hôpital Lyon Sud, Pierre-Bénite, France
| | - Kahina Saker
- Joint Research Unit Hospices Civils de Lyon-bioMerieux S.A., Hôpital Lyon Sud, Pierre-Bénite, France
| | - Priscille Franc
- Joint Research Unit Hospices Civils de Lyon-bioMerieux S.A., Hôpital Lyon Sud, Pierre-Bénite, France
| | - Bouchra Mokdad
- Joint Research Unit Hospices Civils de Lyon-bioMerieux S.A., Hôpital Lyon Sud, Pierre-Bénite, France
| | - Aurore Fleurie
- Joint Research Unit Hospices Civils de Lyon-bioMerieux S.A., Hôpital Lyon Sud, Pierre-Bénite, France
| | - Xavier Lacoux
- R&D - Immunoassay, bioMerieux S.A., Marcy l'Etoile, France
| | - Soizic Daniel
- R&D - Immunoassay, bioMerieux S.A., Marcy l'Etoile, France
| | - Franck Berthier
- R&D - Life Sciences, bioMerieux S.A., Marcy l'Etoile, France
| | - Cécile Barnel
- Joint Research Unit Hospices Civils de Lyon-bioMerieux S.A., Hôpital Lyon Sud, Pierre-Bénite, France
| | - Bruno Pozzetto
- CIRI - Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Lyon, France
- Department of Infectious Agents and Hygiene, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Priest-en-Jarez, France
| | - Jean-Baptiste Fassier
- Department of Occupational Health and Medicine, Hospices Civils de Lyon, Lyon, France
- UMRESTTE (UMR T9405), Université Claude Bernard Lyon 1, Lyon, France
| | - Valérie Dubois
- Etablissement Français du Sang Auvergne Rhône Alpes, Laboratoire HLA de Lyon, Décines, France
| | - Sophia Djebali
- CIRI - Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Lyon, France
| | - Maxence Dubois
- CIRI - Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Lyon, France
| | - Thierry Walzer
- CIRI - Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Lyon, France
| | - Jacqueline Marvel
- CIRI - Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Lyon, France
| | - Karen Brengel-Pesce
- Joint Research Unit Hospices Civils de Lyon-bioMerieux S.A., Hôpital Lyon Sud, Pierre-Bénite, France
| | - Sophie Trouillet-Assant
- CIRI - Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Lyon, France
- Joint Research Unit Hospices Civils de Lyon-bioMerieux S.A., Hôpital Lyon Sud, Pierre-Bénite, France
| |
Collapse
|
7
|
Martín-Galiano AJ, López D. Conservation of HLA Spike Protein Epitopes Supports T Cell Cross-Protection in SARS-CoV-2 Vaccinated Individuals against the Potentially Zoonotic Coronavirus Khosta-2. Int J Mol Sci 2024; 25:6087. [PMID: 38892276 PMCID: PMC11172828 DOI: 10.3390/ijms25116087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Heterologous vaccines, which induce immunity against several related pathogens, can be a very useful and rapid way to deal with new pandemics. In this study, the potential impact of licensed COVID-19 vaccines on cytotoxic and helper cell immune responses against Khosta-2, a novel sarbecovirus that productively infects human cells, was analyzed for the 567 and 41 most common HLA class I and II alleles, respectively. Computational predictions indicated that most of these 608 alleles, covering more than 90% of the human population, contain sufficient fully conserved T-cell epitopes between the Khosta-2 and SARS-CoV-2 spike-in proteins. Ninety percent of these fully conserved peptides for class I and 93% for class II HLA molecules were verified as epitopes recognized by CD8+ or CD4+ T lymphocytes, respectively. These results show a very high correlation between bioinformatic prediction and experimental assays, which strongly validates this study. This immunoinformatics analysis allowed a broader assessment of the alleles that recognize these peptides, a global approach at the population level that is not possible with experimental assays. In summary, these findings suggest that both cytotoxic and helper cell immune protection elicited by currently licensed COVID-19 vaccines should be effective against Khosta-2 virus infection. Finally, by being rapidly adaptable to future coronavirus pandemics, this study has potential public health implications.
Collapse
Affiliation(s)
- Antonio J. Martín-Galiano
- Core Scientific and Technical Units, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain;
| | - Daniel López
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain
| |
Collapse
|
8
|
Andreu-Ballester JC, Galindo-Regal L, Cuéllar C, López-Chuliá F, García-Ballesteros C, Fernández-Murga L, Llombart-Cussac A, Domínguez-Márquez MV. A Low Number of Baselines γδ T Cells Increases the Risk of SARS-CoV-2 Post-Vaccination Infection. Vaccines (Basel) 2024; 12:553. [PMID: 38793803 PMCID: PMC11125751 DOI: 10.3390/vaccines12050553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 05/10/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
Background: The COVID-19 pandemic is the biggest global health problem in the last hundred years. The efficacy of the vaccine to protect against severe disease is estimated to be 70-95% according to the studies carried out, although there are aspects of the immune response to the vaccine that remain unclear. Methods: Humoral and cellular immunity after the administration of three doses of the Pfizer-BioNTech and Oxford AstraZeneca vaccines against SARS-CoV-2 over one year and the appearance of post-vaccination COVID-19 were studied. SARS-CoV-2 IgG and IgA antibodies, αβ and γδ T-cell subsets, and their differentiation stages and apoptosis were analyzed. Results: Anti-SARS-CoV-2 IgG and IgA antibodies showed a progressive increase throughout the duration of the study. This increase was the greatest after the third dose. The highest levels were observed in subjects who had anti-SARS-CoV-2 antibodies prior to vaccination. There was an increase in CD4+ αβ, CD8+ γδ and TEM CD8+ γδ T cells, and a decrease in apoptosis in CD4+ CD8+ and CD56+ αβ and γδ T cells. Post-vaccination SARS-CoV-2 infection was greater than 60%. The symptoms of COVID-19 were very mild and were related to a γδ T cell deficit, specifically CD8+ TEMRA and CD56+ γδ TEM, as well as lower pre-vaccine apoptosis levels. Conclusions: The results unveil the important role of γδ T cells in SARS-CoV-2-vaccine-mediated protection from the disease.
Collapse
Affiliation(s)
- Juan Carlos Andreu-Ballester
- FISABIO Foundation, 46020 Valencia, Spain; (L.G.-R.); (F.L.-C.)
- Parasitic Immunobiology and Immunomodulation Research Group (INMUNOPAR), Complutense University of Madrid, 28040 Madrid, Spain;
| | - Lorena Galindo-Regal
- FISABIO Foundation, 46020 Valencia, Spain; (L.G.-R.); (F.L.-C.)
- Laboratory of Molecular Biology and Research Department, Arnau de Vilanova University Hospital, FISABIO Foundation, 46015 Valencia, Spain;
| | - Carmen Cuéllar
- Parasitic Immunobiology and Immunomodulation Research Group (INMUNOPAR), Complutense University of Madrid, 28040 Madrid, Spain;
- Microbiology and Parasitology Department, Complutense University, 28040 Madrid, Spain
| | - Francisca López-Chuliá
- FISABIO Foundation, 46020 Valencia, Spain; (L.G.-R.); (F.L.-C.)
- Hematology Department, Arnau de Vilanova Hospital, 46015 Valencia, Spain
- Medicine Department, Cardenal Herrera University, 46115 Valencia, Spain
| | - Carlos García-Ballesteros
- Laboratory of Molecular Biology and Research Department, Arnau de Vilanova University Hospital, FISABIO Foundation, 46015 Valencia, Spain;
- Hematology Department, Arnau de Vilanova Hospital, 46015 Valencia, Spain
| | | | | | | |
Collapse
|
9
|
Wells CR, Pandey A, Moghadas SM, Fitzpatrick MC, Singer BH, Galvani AP. Evaluation of Strategies for Transitioning to Annual SARS-CoV-2 Vaccination Campaigns in the United States. Ann Intern Med 2024; 177:609-617. [PMID: 38527289 DOI: 10.7326/m23-2451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND The U.S. Food and Drug Administration has proposed administering annual SARS-CoV-2 vaccines. OBJECTIVE To evaluate the effectiveness of an annual SARS-CoV-2 vaccination campaign, quantify the health and economic benefits of a second dose provided to children younger than 2 years and adults aged 50 years or older, and optimize the timing of a second dose. DESIGN An age-structured dynamic transmission model. SETTING United States. PARTICIPANTS A synthetic population reflecting demographics and contact patterns in the United States. INTERVENTION Vaccination against SARS-CoV-2 with age-specific uptake similar to that of influenza vaccination. MEASUREMENTS Incidence, hospitalizations, deaths, and direct health care cost. RESULTS The optimal timing between the first and second dose delivered to children younger than 2 years and adults aged 50 years or older in an annual vaccination campaign was estimated to be 5 months. In direct comparison with a single-dose campaign, a second booster dose results in 123 869 fewer hospitalizations (95% uncertainty interval [UI], 121 994 to 125 742 fewer hospitalizations) and 5524 fewer deaths (95% UI, 5434 to 5613 fewer deaths), averting $3.63 billion (95% UI, $3.57 billion to $3.69 billion) in costs over a single year. LIMITATIONS Population immunity is subject to degrees of immune evasion for emerging SARS-CoV-2 variants. The model was implemented in the absence of nonpharmaceutical interventions and preexisting vaccine-acquired immunity. CONCLUSION The direct health care costs of SARS-CoV-2, particularly among adults aged 50 years or older, would be substantially reduced by administering a second dose 5 months after the initial dose. PRIMARY FUNDING SOURCE Natural Sciences and Engineering Research Council of Canada, Notsew Orm Sands Foundation, National Institutes of Health, Centers for Disease Control and Prevention, and National Science Foundation.
Collapse
Affiliation(s)
- Chad R Wells
- Center for Infectious Disease Modeling and Analysis, Yale School of Public Health, New Haven, Connecticut (C.R.W., A.P., A.P.G.)
| | - Abhishek Pandey
- Center for Infectious Disease Modeling and Analysis, Yale School of Public Health, New Haven, Connecticut (C.R.W., A.P., A.P.G.)
| | - Seyed M Moghadas
- Agent-Based Modelling Laboratory, York University, Toronto, Ontario, Canada (S.M.M.)
| | - Meagan C Fitzpatrick
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland (M.C.F.)
| | - Burton H Singer
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida (B.H.S.)
| | - Alison P Galvani
- Center for Infectious Disease Modeling and Analysis, Yale School of Public Health, New Haven, Connecticut (C.R.W., A.P., A.P.G.)
| |
Collapse
|
10
|
Zhang N, Wang Z, Li H, Chen K, Wang HS, Shao JB, Jiang SY, Zhai XW, Jiang H. Immunophenotype of lymphocytes and real-world outcome of COVID-19 infection in children with hematology and oncology. BMC Cancer 2024; 24:538. [PMID: 38678181 PMCID: PMC11056048 DOI: 10.1186/s12885-024-12262-1] [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: 09/19/2023] [Accepted: 04/15/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND Patients with immunocompromise were suspected to encounter a high risk for severe coronavirus disease 2019 (COVID-19) infection on early period; however, data is lacking nowadays and immune response remain unclear. METHODS In this retrospective study, internet questionnaire survey and medical records were acquired in pediatric hematology oncology patients. Clinical severity, immunological characteristics, and outcomes were analyzed from December 1, 2022 to January 31, 2023 at the 3rd year of pandemic in China. RESULTS A total of 306 patients were included, with 21 patients (6.9%) asymptomatic, 262 (85.6%) mild severity, 17 (5.6%) moderate severity, 5 (1.6%) severe severity, and 1 (0.3%) critical severity. Seventy-eight (25.5%) patients were on intensive chemotherapy, and 32.0% children were on maintenance chemotherapy. Delays in cancer therapy occurred in 86.7% patients. Univariable analysis revealed active chemotherapy (P < 0.0001), long duration of symptom (P < 0.0001), low lymphocytes count (P = 0.095), low CD3 + and CD8 + T cell count (P = 0.013, P = 0.022), high percentage of CD4 + TCM (P = 0.016), and low percentage of transitional B cells (P = 0.045) were high risk factors for severe COVID-19 infection. Cox regression model showed that the absolute lymphocytes count (P = 0.027) and long duration of symptom (P = 0.002) were the independent factors for severity. Patients with CD8 + dominant and B cell depletion subtype wasn't related with severity, but had higher percentage of CD8 + effector memory T cells (TEM) and terminally differentiated effector memory T cells (TEMRA) (P < 0.001, P < 0.001), and a longer COVID-19 duration (P = 0.045). CONCLUSION The severity was relatively mild in children with immunodeficiencies in the third year of COVID-19 pandemic. Low lymphocyte count and long duration of symptom were the independent risk factors with COVID-19 severity. Delays in cancer care remain a major concern and the long outcome is pending.
Collapse
Affiliation(s)
- Na Zhang
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200040, Shanghai, China
| | - Zhen Wang
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200040, Shanghai, China
| | - Hong Li
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200040, Shanghai, China
| | - Kai Chen
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200040, Shanghai, China
| | - Hong-Sheng Wang
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, China
| | - Jing-Bo Shao
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200040, Shanghai, China
| | - Sha-Yi Jiang
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200040, Shanghai, China
| | - Xiao-Wen Zhai
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, China
| | - Hui Jiang
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200040, Shanghai, China.
| |
Collapse
|
11
|
White CL, Glover MA, Gandhapudi SK, Richards KA, Sant AJ. Flublok Quadrivalent Vaccine Adjuvanted with R-DOTAP Elicits a Robust and Multifunctional CD4 T Cell Response That Is of Greater Magnitude and Functional Diversity Than Conventional Adjuvant Systems. Vaccines (Basel) 2024; 12:281. [PMID: 38543915 PMCID: PMC10975948 DOI: 10.3390/vaccines12030281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/20/2024] [Accepted: 02/29/2024] [Indexed: 04/01/2024] Open
Abstract
It is clear that new approaches are needed to promote broadly protective immunity to viral pathogens, particularly those that are prone to mutation and escape from antibody-mediated immunity. CD4+ T cells, known to target many viral proteins and highly conserved peptide epitopes, can contribute greatly to protective immunity through multiple mechanisms. Despite this potential, CD4+ T cells are often poorly recruited by current vaccine strategies. Here, we have analyzed a promising new adjuvant (R-DOTAP), as well as conventional adjuvant systems AddaVax with or without an added TLR9 agonist CpG, to promote CD4+ T cell responses to the licensed vaccine Flublok containing H1, H3, and HA-B proteins. Our studies, using a preclinical mouse model of vaccination, revealed that the addition of R-DOTAP to Flublok dramatically enhances the magnitude and functionality of CD4+ T cells specific for HA-derived CD4+ T cell epitopes, far outperforming conventional adjuvant systems based on cytokine EliSpot assays and multiparameter flow cytometry. The elicited CD4+ T cells specific for HA-derived epitopes produce IL-2, IFN-γ, IL-4/5, and granzyme B and have multifunctional potential. Hence, R-DOTAP, which has been verified safe by human studies, can offer exciting opportunities as an immune stimulant for next-generation prophylactic recombinant protein-based vaccines.
Collapse
Affiliation(s)
- Chantelle L. White
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (C.L.W.); (M.A.G.); (K.A.R.)
| | - Maryah A. Glover
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (C.L.W.); (M.A.G.); (K.A.R.)
| | - Siva K. Gandhapudi
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky School of Medicine, Lexington, KY 40508, USA;
| | - Katherine A. Richards
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (C.L.W.); (M.A.G.); (K.A.R.)
| | - Andrea J. Sant
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (C.L.W.); (M.A.G.); (K.A.R.)
| |
Collapse
|
12
|
Hsieh MS, Hsu CW, Liao HC, Lin CL, Chiang CY, Chen MY, Liu SJ, Liao CL, Chen HW. SARS-CoV-2 spike-FLIPr fusion protein plus lipidated FLIPr protects against various SARS-CoV-2 variants in hamsters. J Virol 2024; 98:e0154623. [PMID: 38299865 PMCID: PMC10878263 DOI: 10.1128/jvi.01546-23] [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: 10/03/2023] [Accepted: 12/22/2023] [Indexed: 02/02/2024] Open
Abstract
Vaccine-induced mucosal immunity and broad protective capacity against various severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants remain inadequate. Formyl peptide receptor-like 1 inhibitory protein (FLIPr), produced by Staphylococcus aureus, can bind to various Fcγ receptor subclasses. Recombinant lipidated FLIPr (rLF) was previously found to be an effective adjuvant. In this study, we developed a vaccine candidate, the recombinant Delta SARS-CoV-2 spike (rDS)-FLIPr fusion protein (rDS-F), which employs the property of FLIPr binding to various Fcγ receptors. Our study shows that rDS-F plus rLF promotes rDS capture by dendritic cells. Intranasal vaccination of mice with rDS-F plus rLF increases persistent systemic and mucosal antibody responses and CD4/CD8 T-cell responses. Importantly, antibodies induced by rDS-F plus rLF vaccination neutralize Delta, Wuhan, Alpha, Beta, and Omicron strains. Additionally, rDS-F plus rLF provides protective effects against various SARS-CoV-2 variants in hamsters by reducing inflammation and viral loads in the lung. Therefore, rDS-F plus rLF is a potential vaccine candidate to induce broad protective responses against various SARS-CoV-2 variants.IMPORTANCEMucosal immunity is vital for combating pathogens, especially in the context of respiratory diseases like COVID-19. Despite this, most approved vaccines are administered via injection, providing systemic but limited mucosal protection. Developing vaccines that stimulate both mucosal and systemic immunity to address future coronavirus mutations is a growing trend. However, eliciting strong mucosal immune responses without adjuvants remains a challenge. In our study, we have demonstrated that using a recombinant severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike-formyl peptide receptor-like 1 inhibitory protein (FLIPr) fusion protein as an antigen, in combination with recombinant lipidated FLIPr as an effective adjuvant, induced simultaneous systemic and mucosal immune responses through intranasal immunization in mice and hamster models. This approach offered protection against various SARS-CoV-2 strains, making it a promising vaccine candidate for broad protection. This finding is pivotal for future broad-spectrum vaccine development.
Collapse
Affiliation(s)
- Ming-Shu Hsieh
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chia-Wei Hsu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Hung-Chun Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chang-Ling Lin
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chen-Yi Chiang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Mei-Yu Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Shih-Jen Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Len Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Hsin-Wei Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
13
|
Masiá M, de la Rica A, Fernández-González M, García JA, Padilla S, García-Abellán J, Botella Á, Mascarell P, Gutiérrez F. Integrating SARS-CoV-2-specific interferon-γ release assay testing in the evaluation of patients hospitalized with COVID-19. Microbiol Spectr 2023; 11:e0241923. [PMID: 37855635 PMCID: PMC10715100 DOI: 10.1128/spectrum.02419-23] [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: 06/08/2023] [Accepted: 09/06/2023] [Indexed: 10/20/2023] Open
Abstract
IMPORTANCE The cellular immune response is essential in the protection against severe disease in patients with established SARS-CoV-2 infection. The novelty of this study lies in the evaluation of the overall performance of a standardized assay to measure cellular immune response, the SARS-CoV-2-specific interferon-γ release assay (IGRA), in hospitalized patients with severe COVID-19. The SARS-CoV-2 IGRA was shown to accurately classify patients based on disease severity and prognosis, and the study revealed that test performance was not affected by the SARS-CoV-2 variant or control tube results. We identified an assay cut-off point with a high negative predictive value against mortality. The SARS-CoV-2 IGRA in patients hospitalized for COVID-19 may be a useful tool to assess cellular immunity and adopt targeted therapeutic and preventive measures.
Collapse
Affiliation(s)
- Mar Masiá
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Medicine Department, Universidad Miguel Hernández, San Juan de Alicante, Spain
| | - Alba de la Rica
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
- Microbiology Service, Hospital General Universitario de Elche, Alicant, Spain
| | - Marta Fernández-González
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - José Alberto García
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Sergio Padilla
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Medicine Department, Universidad Miguel Hernández, San Juan de Alicante, Spain
| | - Javier García-Abellán
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Medicine Department, Universidad Miguel Hernández, San Juan de Alicante, Spain
| | - Ángela Botella
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
| | - Paula Mascarell
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
| | - Félix Gutiérrez
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Medicine Department, Universidad Miguel Hernández, San Juan de Alicante, Spain
| |
Collapse
|
14
|
El-Saed A, Othman F, Baffoe-Bonnie H, Almulhem R, Matalqah M, Alshammari L, Alshamrani MM. Symptomatic MERS-CoV infection reduces the risk of future COVID-19 disease; a retrospective cohort study. BMC Infect Dis 2023; 23:757. [PMID: 37924004 PMCID: PMC10623690 DOI: 10.1186/s12879-023-08763-2] [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: 04/30/2023] [Accepted: 10/27/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND The general human immune responses similarity against different coronaviruses may reflect some degree of cross-immunity, whereby exposure to one coronavirus may confer partial immunity to another. The aim was to determine whether previous MERS-CoV infection was associated with a lower risk of subsequent COVID-19 disease and its related outcomes. METHODS We conducted a retrospective cohort study among all patients screened for MERS-CoV at a tertiary care hospital in Saudi Arabia between 2012 and early 2020. Both MERS-CoV positive and negative patients were followed up from early 2020 to September 2021 for developing COVID-19 infection confirmed by RT-PCR testing. RESULTS A total of 397 participants followed for an average 15 months during COVID-19 pandemic (4.9 years from MERS-CoV infection). Of the 397 participants, 93 (23.4%) were positive for MERS-CoV at baseline; 61 (65.6%) of the positive cases were symptomatic. Out of 397, 48 (12.1%) participants developed COVID-19 by the end of the follow-up period. Cox regression analysis adjusted for age, gender, and major comorbidity showed a marginally significant lower risk of COVID-19 disease (hazard ratio = 0.533, p = 0.085) and hospital admission (hazard ratio = 0.411, p = 0.061) in patients with positive MERS-CoV. Additionally, the risk of COVID-19 disease was further reduced and became significant in patients with symptomatic MERS-CoV infection (hazard ratio = 0.324, p = 0.034) and hospital admission (hazard ratio = 0.317, p = 0.042). CONCLUSIONS The current findings may indicate a partial cross-immunity, where patients with symptomatic MERS-CoV have a lower risk of future COVID-19 infection and related hospitalization. The present results may need further examination nationally using immunity markers.
Collapse
Affiliation(s)
- Aiman El-Saed
- Infection Prevention and Control Department, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Fatmah Othman
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- Epidemiology and Biostatistics Department, College of Public Health and Health Informatics, King Saud bin Abdul Aziz University for Health Science, Riyadh, Saudi Arabia
| | - Henry Baffoe-Bonnie
- Infection Prevention and Control Department, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Rawabi Almulhem
- Infection Prevention and Control Department, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Muayed Matalqah
- Infection Prevention and Control Department, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Latifah Alshammari
- Infection Prevention and Control Department, King Abdullah Specialist Children Hospital, Riyadh, Saudi Arabia
| | - Majid M Alshamrani
- Infection Prevention and Control Department, King Abdulaziz Medical City, Riyadh, Saudi Arabia.
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia.
| |
Collapse
|
15
|
Domènech-Montoliu S, Puig-Barberà J, Badenes-Marques G, Gil-Fortuño M, Orrico-Sánchez A, Pac-Sa MR, Perez-Olaso O, Sala-Trull D, Sánchez-Urbano M, Arnedo-Pena A. Long COVID Prevalence and the Impact of the Third SARS-CoV-2 Vaccine Dose: A Cross-Sectional Analysis from the Third Follow-Up of the Borriana Cohort, Valencia, Spain (2020-2022). Vaccines (Basel) 2023; 11:1590. [PMID: 37896992 PMCID: PMC10611325 DOI: 10.3390/vaccines11101590] [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: 08/31/2023] [Revised: 09/27/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND In March 2020, a COVID-19 outbreak linked to mass gathering dinners at the Falles Festival in Borriana, Spain, resulted in an estimated attack rate of 42.6% among attendees. METHODS In June 2022, we conducted a cross-sectional follow-up study of 473 adults aged 18 to 64 who attended the dinners at the Falles Festival in 2020, examining the cumulative experience after SARS-CoV-2 infection and vaccination responses. Data included demographic details, lifestyle habits, medical history, infection records, and vaccinations from a population-based vaccine registry. Blood samples were analyzed for SARS-CoV-2 antibodies and cellular immunity. We employed a doubly robust inverse-probability weighting analysis to estimate the booster vaccine dose's impact on long COVID prevalence and symptom count. RESULTS A total of 28.1% of participants met the WHO criteria for long COVID, with older individuals showing higher rates. Long COVID diagnosis was less likely with factors including O blood group, higher occupational status, physical activity, three vaccine doses, strong SARS-CoV-2-S-reactive IFNγ-producing-CD8+ response, and infection during the Omicron period. Increased age, high or low social activity, underlying health conditions, a severe initial COVID episode, and reinfection were associated with higher long COVID likelihood. A booster dose, compared to one or two doses, reduced long COVID risk by 74% (95% CI: 56% to 92%) and symptom count by 55% (95% CI: 32% to 79%). CONCLUSION Long COVID was prevalent in a significant portion of those who contracted COVID-19, underscoring the need for sustained follow-up and therapeutic strategies. Vaccinations, notably the booster dose, had a substantial beneficial effect on long-term infection outcomes, affirming the vaccination's role in mitigating SARS-CoV-2 infection consequences.
Collapse
Affiliation(s)
| | - Joan Puig-Barberà
- Vaccine Research Unit (AIV), Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), 46020 Valencia, Spain;
| | - Gema Badenes-Marques
- Emergency Service, University Hospital de la Plana, 12540 Vila-real, Spain (M.S.-U.)
| | - María Gil-Fortuño
- Microbiology Service, University Hospital de la Plana, 12540 Vila-real, Spain; (M.G.-F.); (O.P.-O.)
| | - Alejandro Orrico-Sánchez
- Vaccine Research Unit (AIV), Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), 46020 Valencia, Spain;
| | - María Rosario Pac-Sa
- Centro de Salud Pública de Castellón, 12003 Castelló de la Plana, Spain; (M.R.P.-S.)
| | - Oscar Perez-Olaso
- Microbiology Service, University Hospital de la Plana, 12540 Vila-real, Spain; (M.G.-F.); (O.P.-O.)
| | - Diego Sala-Trull
- Emergency Service, University Hospital de la Plana, 12540 Vila-real, Spain (M.S.-U.)
| | - Manuel Sánchez-Urbano
- Emergency Service, University Hospital de la Plana, 12540 Vila-real, Spain (M.S.-U.)
| | - Alberto Arnedo-Pena
- Centro de Salud Pública de Castellón, 12003 Castelló de la Plana, Spain; (M.R.P.-S.)
- Department of Health Science, Universidad Pública de Navarra, 31006 Pamplona, Spain
| |
Collapse
|
16
|
Constantin T, Pék T, Horváth Z, Garan D, Szabó AJ. Multisystem inflammatory syndrome in children (MIS-C): Implications for long COVID. Inflammopharmacology 2023; 31:2221-2236. [PMID: 37460909 PMCID: PMC10518292 DOI: 10.1007/s10787-023-01272-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 09/26/2023]
Abstract
The COVID-19 pandemic caused by the coronavirus 2 of the severe acute respiratory syndrome (SARS-CoV-2) has significantly affected people around the world, leading to substantial morbidity and mortality. Although the pandemic has affected people of all ages, there is increasing evidence that children are less susceptible to SARS-CoV-2 infection and are more likely to experience milder symptoms than adults. However, children with COVID-19 can still develop serious complications, such as multisystem inflammatory syndrome in children (MIS-C). This narrative review of the literature provides an overview of the epidemiology and immune pathology of SARS-CoV-2 infection and MIS-C in children. The review also examines the genetics of COVID-19 and MIS-C in children, including the genetic factors that can influence the susceptibility and severity of the diseases and their implications for personalized medicine and vaccination strategies. By examining current evidence and insights from the literature, this review aims to contribute to the development of effective prevention and treatment strategies for COVID-19, MIS-C, and long COVID syndromes in children.
Collapse
Affiliation(s)
- Tamás Constantin
- Department of Pediatrics, Semmelweis University, Tűzoltó u. 7-9., Budapest, 1094, Hungary.
| | - Tamás Pék
- Department of Pediatrics, Semmelweis University, Tűzoltó u. 7-9., Budapest, 1094, Hungary
| | - Zsuzsanna Horváth
- Department of Pediatrics, Semmelweis University, Tűzoltó u. 7-9., Budapest, 1094, Hungary
| | - Diána Garan
- Department of Pediatrics, Semmelweis University, Tűzoltó u. 7-9., Budapest, 1094, Hungary
| | - Attila J Szabó
- Department of Pediatrics, Semmelweis University, Tűzoltó u. 7-9., Budapest, 1094, Hungary
| |
Collapse
|
17
|
Hollstein MM, Dierks S, Schön MP, Bergmann A, Abratis A, Eidizadeh A, Kaltenbach S, Schanz J, Groß U, Leha A, Kröger A, Andag R, Zautner AE, Fischer A, Erpenbeck L, Schnelle M. Humoral and cellular immune responses in fully vaccinated individuals with or without SARS-CoV-2 breakthrough infection: Results from the CoV-ADAPT cohort. J Med Virol 2023; 95:e29122. [PMID: 37787583 DOI: 10.1002/jmv.29122] [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: 03/31/2023] [Revised: 08/15/2023] [Accepted: 09/13/2023] [Indexed: 10/04/2023]
Abstract
Despite recent advances in prophylactic vaccination, SARS-CoV-2 infections continue to cause significant morbidity. A better understanding of immune response differences between vaccinated individuals with and without later SARS-CoV-2 breakthrough infection is urgently needed. CoV-ADAPT is a prospective long-term study comparing humoral (anti-spike-RBD-IgG, neutralization capacity, avidity) and cellular (spike-induced T-cell interferon-γ [IFN-γ] release) immune responses in individuals vaccinated against SARS-CoV-2 at four different time points (three before and one after third vaccination). In this cohort study, 62 fully vaccinated individuals presented with SARS-CoV-2 breakthrough infections vs 151 without infection 3-7 months following third vaccination. Breakthrough infections significantly increased anti-spike-RBD-IgG (p < 0.01), but not spike-directed T-cell IFN-γ release (TC) or antibody avidity. Despite comparable surrogate neutralization indices, the functional neutralization capacity against SARS-CoV-2-assessed via a tissue culture-based assay-was significantly higher following breakthrough vs no breakthrough infection. Anti-spike-RBD-IgG and antibody avidity decreased with age (p < 0.01) and females showed higher anti-spike-RBD-IgG (p < 0.01), and a tendency towards higher antibody avidity (p = 0.051). The association between humoral and cellular immune responses previously reported at various time points was lost in subjects after breakthrough infections (p = 0.807). Finally, a machine-learning approach based on our large immunological dataset (a total of 49 variables) from different time points was unable to predict breakthrough infections (area under the curve: 0.55). In conclusion, distinct differences in humoral vs cellular immune responses in fully vaccinated individuals with or without breakthrough infection could be demonstrated. Breakthrough infections predominantly drive the humoral response without boosting the cellular component. Breakthrough infections could not be predicted based on immunological data, which indicates a superior role of environmental factors (e.g., virus exposure) in individualized risk assessment.
Collapse
Affiliation(s)
- Moritz M Hollstein
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
| | - Sascha Dierks
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
- Interdisciplinary UMG Laboratory, University Medical Center Göttingen, Göttingen, Germany
| | - Michael P Schön
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
- Lower Saxony Institute of Occupational Dermatology, University Medical Center Göttingen, Göttingen, Germany
| | - Armin Bergmann
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
| | - Anna Abratis
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
- Interdisciplinary UMG Laboratory, University Medical Center Göttingen, Göttingen, Germany
| | - Abass Eidizadeh
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
- Interdisciplinary UMG Laboratory, University Medical Center Göttingen, Göttingen, Germany
| | - Sarah Kaltenbach
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
- Interdisciplinary UMG Laboratory, University Medical Center Göttingen, Göttingen, Germany
| | - Julie Schanz
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Uwe Groß
- Interdisciplinary UMG Laboratory, University Medical Center Göttingen, Göttingen, Germany
- Institute of Medical Microbiology and Virology, University Medical Center Göttingen, Göttingen, Germany
| | - Andreas Leha
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| | - Andrea Kröger
- Institute of Medical Microbiology and Hospital Hygiene, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Research Group Innate Immunity and Infection, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Reiner Andag
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
- Interdisciplinary UMG Laboratory, University Medical Center Göttingen, Göttingen, Germany
| | - Andreas E Zautner
- Institute of Medical Microbiology and Virology, University Medical Center Göttingen, Göttingen, Germany
- Institute of Medical Microbiology and Hospital Hygiene, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Center for Health and Medical Prevention (CHaMP), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Andreas Fischer
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
- Interdisciplinary UMG Laboratory, University Medical Center Göttingen, Göttingen, Germany
| | - Luise Erpenbeck
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
- Department of Dermatology, University of Münster, Münster, Germany
| | - Moritz Schnelle
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
- Interdisciplinary UMG Laboratory, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
18
|
Föhse K, Geckin B, Zoodsma M, Kilic G, Liu Z, Röring RJ, Overheul GJ, van de Maat J, Bulut O, Hoogerwerf JJ, Ten Oever J, Simonetti E, Schaal H, Adams O, Müller L, Ostermann PN, van de Veerdonk FL, Joosten LAB, Haagmans BL, van Crevel R, van Rij RP, GeurtsvanKessel C, de Jonge MI, Li Y, Domínguez-Andrés J, Netea MG. The impact of BNT162b2 mRNA vaccine on adaptive and innate immune responses. Clin Immunol 2023; 255:109762. [PMID: 37673225 DOI: 10.1016/j.clim.2023.109762] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/04/2023] [Accepted: 09/02/2023] [Indexed: 09/08/2023]
Abstract
The mRNA-based BNT162b2 protects against severe disease and mortality caused by SARS-CoV-2 via induction of specific antibody and T-cell responses. Much less is known about its broad effects on immune responses against other pathogens. Here, we investigated the adaptive immune responses induced by BNT162b2 vaccination against various SARS-CoV-2 variants and its effects on the responsiveness of immune cells upon stimulation with heterologous stimuli. BNT162b2 vaccination induced effective humoral and cellular immunity against SARS-CoV-2 that started to wane after six months. We also observed long-term transcriptional changes in immune cells after vaccination. Additionally, vaccination with BNT162b2 modulated innate immune responses as measured by inflammatory cytokine production after stimulation - higher IL-1/IL-6 release and decreased IFN-α production. Altogether, these data expand our knowledge regarding the overall immunological effects of this new class of vaccines and underline the need for additional studies to elucidate their effects on both innate and adaptive immune responses.
Collapse
Affiliation(s)
- Konstantin Föhse
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Büsra Geckin
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Martijn Zoodsma
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; TWINCORE, A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Gizem Kilic
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Zhaoli Liu
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; TWINCORE, A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Rutger J Röring
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Gijs J Overheul
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Josephine van de Maat
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ozlem Bulut
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jacobien J Hoogerwerf
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jaap Ten Oever
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Elles Simonetti
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Heiner Schaal
- Institute of Virology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Ortwin Adams
- Institute of Virology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Lisa Müller
- Institute of Virology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Philipp Niklas Ostermann
- Institute of Virology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Frank L van de Veerdonk
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Bart L Haagmans
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Reinout van Crevel
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ronald P van Rij
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Marien I de Jonge
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Yang Li
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; TWINCORE, A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Jorge Domínguez-Andrés
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
19
|
Albiol N, Lynton-Pons E, Aso O, Moga E, Vidal S, Gómez-Pérez L, Santiago JA, Triquell M, Roch N, Lázaro E, González I, López-Contreras J, Esquirol A, Sierra J, Martino R, García-Cadenas I. mRNA-1273 SARS-CoV-2 vaccine in recently transplanted allogeneic hematopoietic cell transplant recipients: Dynamics of cellular and humoral immune responses and booster effect. Leuk Res 2023; 132:107347. [PMID: 37356281 PMCID: PMC10284722 DOI: 10.1016/j.leukres.2023.107347] [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: 03/09/2023] [Revised: 06/18/2023] [Accepted: 06/19/2023] [Indexed: 06/27/2023]
Abstract
Allogeneic hematopoietic stem cell transplant (HCT) recipients are at high risk of severe COVID-19 despite vaccination. Little is known about cellular response to SARS-CoV-2 vaccine in this population, especially in recently transplanted patients (RTP). In this single-center study we examined cellular and humoral response to the mRNA-1273 (Spikevax®) vaccine in recently transplanted patients (RTP, n = 49), and compared them to long-term transplanted patients (LTTP, n = 19) and healthy controls (n = 20) at three different timepoints: one and three months after the second dose (T1 and T2, respectively, 28 days apart), and one month after the third dose (T3). Controls did not receive a third dose. RTPs showed lower IgG anti-S1 titers than healthy controls at both T1 (mean 0.50 vs 0.94 arbitrary units -AU-, p < 0.0001) and T2 (0.37 vs 0.79 AU, p < 0.0001). They also presented lower titers than LTTPs at T1 (0.50 vs 0.66, p = 0.01), but no differences at T2 (0.37 vs 0.40 AU, p = 0.55). The rate of positive T-cell responses was lower in RTPs than in controls at both T1 and T2 (61.2 % vs 95 %, p = 0.007; 59.2 % vs 100 %, p = 0.001, respectively), but without statistically significant differences between transplanted groups. At T3 no differences were seen between RTPs and LTTPs as well, neither in IgG antibodies (p = 0.82) nor in cellular responses (p = 0.15), although a third dose increased the rate of positive cellular and humoral responses in approximately 50 % of recently transplanted patients. However, active immunosuppressive treatment severely diminished their chances to produce an adequate response.
Collapse
Affiliation(s)
- Nil Albiol
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Carrer del Mas Casanovas 90, 08041 Barcelona, Spain; Josep Carreras Leukaemia Research Institute, Carrer de Sant Quintí 77-79, 08041 Barcelona, Spain; Institut d'Investigació Biomèdica (IIB) Sant Pau, Carrer de Sant Quintí 77-79, 08041 Barcelona, Spain; Autonomous University of Barcelona, Carrer de Sant Antoni Maria Claret 167, 08025 Barcelona, Spain; Catalan Institute of Oncology (ICO), Hospital Universitari Doctor Josep Trueta, 17007 Girona, Spain.
| | - Elionor Lynton-Pons
- Institut d'Investigació Biomèdica (IIB) Sant Pau, Carrer de Sant Quintí 77-79, 08041 Barcelona, Spain; Autonomous University of Barcelona, Carrer de Sant Antoni Maria Claret 167, 08025 Barcelona, Spain; Immunology Department, Hospital de la Santa Creu i Sant Pau, Carrer del Mas Casanovas 90, 08041 Barcelona, Spain
| | - Olga Aso
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Carrer del Mas Casanovas 90, 08041 Barcelona, Spain
| | - Esther Moga
- Josep Carreras Leukaemia Research Institute, Carrer de Sant Quintí 77-79, 08041 Barcelona, Spain; Institut d'Investigació Biomèdica (IIB) Sant Pau, Carrer de Sant Quintí 77-79, 08041 Barcelona, Spain; Autonomous University of Barcelona, Carrer de Sant Antoni Maria Claret 167, 08025 Barcelona, Spain; Immunology Department, Hospital de la Santa Creu i Sant Pau, Carrer del Mas Casanovas 90, 08041 Barcelona, Spain
| | - Silvia Vidal
- Institut d'Investigació Biomèdica (IIB) Sant Pau, Carrer de Sant Quintí 77-79, 08041 Barcelona, Spain; Autonomous University of Barcelona, Carrer de Sant Antoni Maria Claret 167, 08025 Barcelona, Spain; Immunology Department, Hospital de la Santa Creu i Sant Pau, Carrer del Mas Casanovas 90, 08041 Barcelona, Spain
| | - Lucía Gómez-Pérez
- Hematology Department, Hospital del Mar - Parc de Salut Mar, Passeig Marítim de la Barceloneta 25-29, 08003 Barcelona, Spain
| | - Jose Alejandre Santiago
- Institut d'Investigació Biomèdica (IIB) Sant Pau, Carrer de Sant Quintí 77-79, 08041 Barcelona, Spain; Immunology Department, Hospital de la Santa Creu i Sant Pau, Carrer del Mas Casanovas 90, 08041 Barcelona, Spain
| | - Mercè Triquell
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Carrer del Mas Casanovas 90, 08041 Barcelona, Spain
| | - Nerea Roch
- Division of Infectious Diseases, Hospital de la Santa Creu i Sant Pau, Carrer del Mas Casanovas 90, 08041 Barcelona, Spain
| | - Elisabeth Lázaro
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Carrer del Mas Casanovas 90, 08041 Barcelona, Spain
| | - Iria González
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Carrer del Mas Casanovas 90, 08041 Barcelona, Spain; Autonomous University of Barcelona, Carrer de Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - Joaquín López-Contreras
- Institut d'Investigació Biomèdica (IIB) Sant Pau, Carrer de Sant Quintí 77-79, 08041 Barcelona, Spain; Autonomous University of Barcelona, Carrer de Sant Antoni Maria Claret 167, 08025 Barcelona, Spain; Division of Infectious Diseases, Hospital de la Santa Creu i Sant Pau, Carrer del Mas Casanovas 90, 08041 Barcelona, Spain
| | - Albert Esquirol
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Carrer del Mas Casanovas 90, 08041 Barcelona, Spain; Josep Carreras Leukaemia Research Institute, Carrer de Sant Quintí 77-79, 08041 Barcelona, Spain; Institut d'Investigació Biomèdica (IIB) Sant Pau, Carrer de Sant Quintí 77-79, 08041 Barcelona, Spain; Autonomous University of Barcelona, Carrer de Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - Jorge Sierra
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Carrer del Mas Casanovas 90, 08041 Barcelona, Spain; Josep Carreras Leukaemia Research Institute, Carrer de Sant Quintí 77-79, 08041 Barcelona, Spain; Institut d'Investigació Biomèdica (IIB) Sant Pau, Carrer de Sant Quintí 77-79, 08041 Barcelona, Spain; Autonomous University of Barcelona, Carrer de Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - Rodrigo Martino
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Carrer del Mas Casanovas 90, 08041 Barcelona, Spain; Josep Carreras Leukaemia Research Institute, Carrer de Sant Quintí 77-79, 08041 Barcelona, Spain; Institut d'Investigació Biomèdica (IIB) Sant Pau, Carrer de Sant Quintí 77-79, 08041 Barcelona, Spain; Autonomous University of Barcelona, Carrer de Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - Irene García-Cadenas
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Carrer del Mas Casanovas 90, 08041 Barcelona, Spain; Josep Carreras Leukaemia Research Institute, Carrer de Sant Quintí 77-79, 08041 Barcelona, Spain; Institut d'Investigació Biomèdica (IIB) Sant Pau, Carrer de Sant Quintí 77-79, 08041 Barcelona, Spain; Autonomous University of Barcelona, Carrer de Sant Antoni Maria Claret 167, 08025 Barcelona, Spain.
| |
Collapse
|
20
|
Chen M, Wang J, Yuan M, Long M, Sun Y, Wang S, Luo W, Zhou Y, Zhang W, Jiang W, Chao J. AT2 cell-derived IgA trapped by the extracellular matrix in silica-induced pulmonary fibrosis. Int Immunopharmacol 2023; 122:110545. [PMID: 37390644 DOI: 10.1016/j.intimp.2023.110545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/14/2023] [Accepted: 06/18/2023] [Indexed: 07/02/2023]
Abstract
Pulmonary fibrosis is an interstitial lung disease caused by various factors such as exposure to workplace environmental contaminants, drugs, or X-rays. Epithelial cells are among the driving factors of pulmonary fibrosis. Immunoglobulin A (IgA), traditionally thought to be secreted by B cells, is an important immune factor involved in respiratory mucosal immunity. In the current study, we found that lung epithelial cells are involved in IgA secretion, which, in turn, promotes pulmonary fibrosis. Spatial transcriptomics and single-cell sequencing suggest that Igha transcripts were highly expressed in the fibrotic lesion areas of lungs from silica-treated mice. Reconstruction of B-cell receptor (BCR) sequences revealed a new cluster of AT2-like epithelial cells with a shared BCR and high expression of genes related to IgA production. Furthermore, the secretion of IgA by AT2-like cells was trapped by the extracellular matrix and aggravated pulmonary fibrosis by activating fibroblasts. Targeted blockade of IgA secretion by pulmonary epithelial cells may be a potential strategy for treating pulmonary fibrosis.
Collapse
Affiliation(s)
- Mengling Chen
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Jing Wang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Mengqin Yuan
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, China
| | - Min Long
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, China
| | - Yuheng Sun
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Sha Wang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Wei Luo
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Yun Zhou
- Department of Health Management, School of Health Science, West Yunnan University of Applied Sciences, Dali, Yunnan, China
| | - Wei Zhang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Wei Jiang
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, China.
| | - Jie Chao
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China; School of Medicine, Xizang Minzu University, Xianyang, Shanxi, China.
| |
Collapse
|
21
|
Muik A, Lui BG, Quandt J, Diao H, Fu Y, Bacher M, Gordon J, Toker A, Grosser J, Ozhelvaci O, Grikscheit K, Hoehl S, Kohmer N, Lustig Y, Regev-Yochay G, Ciesek S, Beguir K, Poran A, Vogler I, Türeci Ö, Sahin U. Progressive loss of conserved spike protein neutralizing antibody sites in Omicron sublineages is balanced by preserved T cell immunity. Cell Rep 2023; 42:112888. [PMID: 37527039 DOI: 10.1016/j.celrep.2023.112888] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 03/27/2023] [Accepted: 07/13/2023] [Indexed: 08/03/2023] Open
Abstract
Evolution of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant has led to the emergence of sublineages with different patterns of neutralizing antibody evasion. We report that Omicron BA.4/BA.5 breakthrough infection of individuals immunized with SARS-CoV-2 wild-type-strain-based mRNA vaccines results in a boost of Omicron BA.4.6, BF.7, BQ.1.1, and BA.2.75 neutralization but does not efficiently boost BA.2.75.2, XBB, or XBB.1.5 neutralization. In silico analyses showed that the Omicron spike glycoprotein lost most neutralizing B cell epitopes, especially in sublineages BA.2.75.2, XBB, and XBB.1.5. In contrast, T cell epitopes are conserved across variants including XBB.1.5. T cell responses of mRNA-vaccinated, SARS-CoV-2-naive individuals against the wild-type strain, Omicron BA.1, and BA.4/BA.5 were comparable, suggesting that T cell immunity against recent sublineages including XBB.1.5 may remain largely unaffected. While some Omicron sublineages effectively evade B cell immunity, spike-protein-specific T cell immunity, due to the nature of polymorphic cell-mediated immune responses, may continue to contribute to prevention/limitation of severe COVID-19 manifestation.
Collapse
Affiliation(s)
| | | | | | - Huitian Diao
- BioNTech US, 40 Erie Street, Cambridge, MA 02139, USA
| | - Yunguan Fu
- InstaDeep, Ltd., 5 Merchant Square, London W2 1AY, UK
| | - Maren Bacher
- BioNTech, An der Goldgrube 12, 55131 Mainz, Germany
| | | | - Aras Toker
- BioNTech, An der Goldgrube 12, 55131 Mainz, Germany
| | | | | | - Katharina Grikscheit
- Institute for Medical Virology, University Hospital, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany
| | - Sebastian Hoehl
- Institute for Medical Virology, University Hospital, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany
| | - Niko Kohmer
- Institute for Medical Virology, University Hospital, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany
| | - Yaniv Lustig
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel; Central Virology Laboratory, Public Health Services, Ministry of Health, Tel-Hashomer, Ramat Gan, Israel
| | - Gili Regev-Yochay
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel; SPRI-Sheba Pandemic Preparedness Research Institute, Sheba Medical Center Tel Hashomer, Ramat Gan, Israel
| | - Sandra Ciesek
- Institute for Medical Virology, University Hospital, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany; DZIF - German Centre for Infection Research, External Partner Site, 60596 Frankfurt am Main, Germany
| | - Karim Beguir
- InstaDeep, Ltd., 5 Merchant Square, London W2 1AY, UK
| | - Asaf Poran
- BioNTech US, 40 Erie Street, Cambridge, MA 02139, USA
| | | | - Özlem Türeci
- BioNTech, An der Goldgrube 12, 55131 Mainz, Germany; HI-TRON - Helmholtz Institute for Translational Oncology Mainz by DKFZ, Obere Zahlbacherstr. 63, 55131 Mainz, Germany
| | - Ugur Sahin
- BioNTech, An der Goldgrube 12, 55131 Mainz, Germany; TRON gGmbH - Translational Oncology at the University Medical Center of the Johannes Gutenberg University, Freiligrathstraße 12, 55131 Mainz, Germany.
| |
Collapse
|
22
|
Zhu D, Wu T, Yu X, Chen Y, Zhou T, Liu Y, Liu L, Min Z. Associations between laboratory variables and clinical features in patients hospitalized with COVID-19 after non-mRNA vaccination in China: A cross-sectional study. Heliyon 2023; 9:e18167. [PMID: 37539143 PMCID: PMC10393604 DOI: 10.1016/j.heliyon.2023.e18167] [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: 12/04/2022] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 08/05/2023] Open
Abstract
Objectives Based on the data during the outbreak of COVID-19 in Wuxi city in China, we explored the relationship between laboratory variables and clinical features in patients hospitalized with COVID-19 after non-mRNA vaccination, and attempted to identify the significant impact of vaccination and COVID-19 infection on humans. Methods A retrospective observational cohort study was carried out. Patients who received non-mRNA COVID-19 vaccines and were hospitalized with COVID-19 between June 28, 2022, and July 24, 2022 were included. The correlation between different vaccine statuses, the time to negative PCR test, and biochemical parameters were investigated. Results All patients had a mild COVID-19 disease. The number of vaccine doses exerted no effects on the time to negative PCR test (P = 0.559). No differences were evident among inactivated, adenoviral-vectored, and recombinant subunit vaccines in the time to negative PCR test.Patients who just received one dose had significantly lower blood glucose levels than those who received three doses (P = 0.024), whereas two doses had no effect on blood glucose levels (one dose vs. two doses, P = 0.223; two doses vs. three doses, P = 0.457).Body temperature (β = 0.168, P = 0.011) and the percentage of lymphocytes (β = -0.219, P = 0.001) were substantially correlated with the time to COVID-19 negative PCR test. The prolonged stay was linked to a rise in GOT that fell within the usual range (P = 0.025).The percentage of lymphocytes (P = 0.007) and serum potassium (P = 0.004) were concordant with the marked change in body temperature. Conclusions The dose and type of vaccination had no effect on the time to COVID-19 negative PCR test in patients with mild COVID-19. Comparing the first dose with the booster dose, the blood glucose levels increased within the normal range. The period at which the COVID-19 nucleic acid turned negative correlated with body temperature, the proportion of lymphocytes, GOT, and serum potassium.
Collapse
|
23
|
Tsoutsoura P, Xagas E, Roussos S, Hatzakis A, Gourzi P, Boletis IN, Marinaki S. Assessment of mRNA Vaccine Immunogenicity in Solid Organ Transplant Recipients. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1075. [PMID: 37374279 DOI: 10.3390/medicina59061075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023]
Abstract
Background and Objectives: Solid organ transplant (SOT) recipients have a higher risk of suffering from severe Coronavirus (COVID-19) compared to the general population. Studies have shown impaired immunogenicity of mRNA vaccines in this high-risk population; thus, SOT recipients have been prioritized globally for primary and booster doses. Materials and Methods: We analyzed 144 SOT recipients who had previously received two doses of BNT162b2 or mRNA1273 vaccine, and who were subsequently vaccinated with a booster dose of the mRNA1273 vaccine. Humoral and cellular immune responses were measured 1 and 3 months after the second dose, and 1 month after the third dose. Results: One month after the second dose, 33.6% (45/134) of patients displayed a positive antibody response with a median (25th, 75th) antibody titer of 9 (7, 161) AU/mL. Three months after the second dose, 41.8% (56/134) tested positive with a median (25th, 75th) antibody titer of 18 (7, 251) AU/mL. After the booster dose, the seropositivity rate increased to 69.4% (93/134), with a median (25th, 75th) titer of 966 (10, 8027) AU/mL. The specific SARS-CoV-2 T-cell response was assessed in 44 randomly selected recipients 3 months after the second dose, and 11.4% (5/44) of them had a positive response. Following the third dose, 42% (21/50) tested positive. Side effects after the third dose were mild, with pain at the injection site being the most frequent adverse effect, reported by 73.4% of the recipients. Conclusion: Our study shows a mild delayed increase in antibody titer, three months after primary vaccination compared to one month after. It also shows a robust augmentation of humoral and specific T-cell responses after the booster dose, as well as the safety and tolerability of the mRNA vaccines in SOT recipients.
Collapse
Affiliation(s)
- Paraskevi Tsoutsoura
- Clinic of Nephrology and Renal Transplantation, Laiko General Hospital, Medical School of Athens, National and Kapodistrian University, 11527 Athens, Greece
| | - Efstathios Xagas
- Clinic of Nephrology and Renal Transplantation, Laiko General Hospital, Medical School of Athens, National and Kapodistrian University, 11527 Athens, Greece
| | - Sotirios Roussos
- Department of Hygiene, Epidemiology & Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Angelos Hatzakis
- Department of Hygiene, Epidemiology & Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Polyxeni Gourzi
- Molecular Immunopathology and Histocompatibility Unit, Onassis Cardiac Surgery Center, 17674 Athens, Greece
| | - Ioannis N Boletis
- Clinic of Nephrology and Renal Transplantation, Laiko General Hospital, Medical School of Athens, National and Kapodistrian University, 11527 Athens, Greece
| | - Smaragdi Marinaki
- Clinic of Nephrology and Renal Transplantation, Laiko General Hospital, Medical School of Athens, National and Kapodistrian University, 11527 Athens, Greece
| |
Collapse
|
24
|
Brisotto G, Montico M, Turetta M, Zanussi S, Cozzi MR, Vettori R, Boschian Boschin R, Vinante L, Matrone F, Revelant A, Palazzari E, Innocente R, Fanetti G, Gerratana L, Garutti M, Lisanti C, Bolzonello S, Nicoloso MS, Steffan A, Muraro E. Integration of Cellular and Humoral Immune Responses as an Immunomonitoring Tool for SARS-CoV-2 Vaccination in Healthy and Fragile Subjects. Viruses 2023; 15:1276. [PMID: 37376576 DOI: 10.3390/v15061276] [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: 05/12/2023] [Revised: 05/26/2023] [Accepted: 05/28/2023] [Indexed: 06/29/2023] Open
Abstract
Cellular and humoral immunity are both required for SARS-CoV-2 infection recovery and vaccine efficacy. The factors affecting mRNA vaccination-induced immune responses, in healthy and fragile subjects, are still under investigation. Thus, we monitored the vaccine-induced cellular and humoral immunity in healthy subjects and cancer patients after vaccination to define whether a different antibody titer reflected similar rates of cellular immune responses and if cancer has an impact on vaccination efficacy. We found that higher titers of antibodies were associated with a higher probability of positive cellular immunity and that this greater immune response was correlated with an increased number of vaccination side effects. Moreover, active T-cell immunity after vaccination was associated with reduced antibody decay. The vaccine-induced cellular immunity appeared more likely in healthy subjects rather than in cancer patients. Lastly, after boosting, we observed a cellular immune conversion in 20% of subjects, and a strong correlation between pre- and post-boosting IFN-γ levels, while antibody levels did not display a similar association. Finally, our data suggested that integrating humoral and cellular immune responses could allow the identification of SARS-CoV-2 vaccine responders and that T-cell responses seem more stable over time compared to antibodies, especially in cancer patients.
Collapse
Affiliation(s)
- Giulia Brisotto
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Marcella Montico
- Clinical Trial Office, Scientific Direction, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Matteo Turetta
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Stefania Zanussi
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Maria Rita Cozzi
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Roberto Vettori
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Romina Boschian Boschin
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Lorenzo Vinante
- Division of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Fabio Matrone
- Division of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Alberto Revelant
- Division of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Elisa Palazzari
- Division of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Roberto Innocente
- Division of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Giuseppe Fanetti
- Division of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Lorenzo Gerratana
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Mattia Garutti
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Camilla Lisanti
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Silvia Bolzonello
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Milena Sabrina Nicoloso
- Molecular Oncology Unit, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Agostino Steffan
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Elena Muraro
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| |
Collapse
|
25
|
Savchenko AA, Kudryavtsev IV, Isakov DV, Sadowski IS, Belenyuk VD, Borisov AG. Recombinant Human Interleukin-2 Corrects NK Cell Phenotype and Functional Activity in Patients with Post-COVID Syndrome. Pharmaceuticals (Basel) 2023; 16:ph16040537. [PMID: 37111294 PMCID: PMC10144656 DOI: 10.3390/ph16040537] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/31/2023] [Accepted: 04/01/2023] [Indexed: 04/07/2023] Open
Abstract
Post-COVID syndrome develops in 10–20% of people who have recovered from COVID-19 and it is characterized by impaired function of the nervous, cardiovascular, and immune systems. Previously, it was found that patients who recovered from infection with the SARS-CoV-2 virus had a decrease in the number and functional activity of NK cells. The aim of the study was to assess the effectiveness of recombinant human IL-2 (rhIL-2) administered to correct NK cell phenotype and functional activity in patients with post-COVID syndrome. Patients were examined after 3 months for acute COVID-19 of varying severity. The phenotype of the peripheral blood NK cells was studied by flow cytometry. It was found that disturbances in the cell subset composition in patients with post-COVID syndrome were characterized by low levels of mature (p = 0.001) and cytotoxic NK cells (p = 0.013), with increased release of immature NK cells (p = 0.023). Functional deficiency of NK cells in post-COVID syndrome was characterized by lowered cytotoxic activity due to the decreased count of CD57+ (p = 0.001) and CD8+ (p < 0.001) NK cells. In the treatment of patients with post-COVID syndrome with recombinant IL-2, peripheral blood NK cell count and functional potential were restored. In general, the effectiveness of using rhIL-2 in treatment of post-COVID syndrome has been proven in patients with low levels of NK cells.
Collapse
Affiliation(s)
- Andrei A. Savchenko
- Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia
| | - Igor V. Kudryavtsev
- Institute of Experimental Medicine, 197376 St. Petersburg, Russia
- School of Biomedicine, Far Eastern Federal University, 690922 Vladivostok, Russia
| | - Dmitry V. Isakov
- Institute of Experimental Medicine, Pavlov First St. Petersburg State Medical University of the Russian Federation Ministry of Healthcare, 197022 St. Petersburg, Russia
| | - Ivan S. Sadowski
- Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia
| | - Vasily D. Belenyuk
- Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia
| | - Alexandr G. Borisov
- Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia
| |
Collapse
|
26
|
Körber N, Holzmann-Littig C, Wilkens G, Liao BH, Werz ML, Platen L, Cheng CC, Tellenbach M, Kappler V, Lehner V, Mijočević H, Christa C, Assfalg V, Heemann U, Schmaderer C, Protzer U, Braunisch MC, Bauer T, Renders L. Comparable cellular and humoral immunity upon homologous and heterologous COVID-19 vaccination regimens in kidney transplant recipients. Front Immunol 2023; 14:1172477. [PMID: 37063863 PMCID: PMC10102365 DOI: 10.3389/fimmu.2023.1172477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 03/21/2023] [Indexed: 04/03/2023] Open
Abstract
BackgroundKidney transplant recipients (KTRs) are at high risk for a severe course of coronavirus disease 2019 (COVID-19); thus, effective vaccination is critical. However, the achievement of protective immunogenicity is hampered by immunosuppressive therapies. We assessed cellular and humoral immunity and breakthrough infection rates in KTRs vaccinated with homologous and heterologous COVID-19 vaccination regimens.MethodWe performed a comparative in-depth analysis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)–specific T-cell responses using multiplex Fluorospot assays and SARS-CoV-2-specific neutralizing antibodies (NAbs) between three-times homologously (n = 18) and heterologously (n = 8) vaccinated KTRs.ResultsWe detected SARS-CoV-2-reactive T cells in 100% of KTRs upon third vaccination, with comparable frequencies, T-cell expression profiles, and relative interferon γ and interleukin 2 production per single cell between homologously and heterologously vaccinated KTRs. SARS-CoV-2-specific NAb positivity rates were significantly higher in heterologously (87.5%) compared to homologously vaccinated (50.0%) KTRs (P < 0.0001), whereas the magnitudes of NAb titers were comparable between both subcohorts after third vaccination. SARS-CoV-2 breakthrough infections occurred in equal numbers in homologously (38.9%) and heterologously (37.5%) vaccinated KTRs with mild-to-moderate courses of COVID-19.ConclusionOur data support a more comprehensive assessment of not only humoral but also cellular SARS-CoV-2-specific immunity in KTRs to provide an in-depth understanding about the COVID-19 vaccine–induced immune response in a transplant setting.
Collapse
Affiliation(s)
- Nina Körber
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
- *Correspondence: Nina Körber,
| | - Christopher Holzmann-Littig
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
- Technical University of Munich (TUM) Medical Education Center, School of Medicine, Technical University of Munich, Munich, Germany
| | - Gesa Wilkens
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
| | - Bo-Hung Liao
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Maia L. Werz
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Louise Platen
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Cho-Chin Cheng
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Myriam Tellenbach
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Verena Kappler
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Viktor Lehner
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Hrvoje Mijočević
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Catharina Christa
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Volker Assfalg
- Department of Surgery, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Uwe Heemann
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Christoph Schmaderer
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Ulrike Protzer
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Matthias C. Braunisch
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Tanja Bauer
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Lutz Renders
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| |
Collapse
|
27
|
Harne R, Williams B, Abdelaal HFM, Baldwin SL, Coler RN. SARS-CoV-2 infection and immune responses. AIMS Microbiol 2023; 9:245-276. [PMID: 37091818 PMCID: PMC10113164 DOI: 10.3934/microbiol.2023015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 03/14/2023] [Accepted: 03/21/2023] [Indexed: 04/03/2023] Open
Abstract
The recent pandemic caused by the SARS-CoV-2 virus continues to be an enormous global challenge faced by the healthcare sector. Availability of new vaccines and drugs targeting SARS-CoV-2 and sequelae of COVID-19 has given the world hope in ending the pandemic. However, the emergence of mutations in the SARS-CoV-2 viral genome every couple of months in different parts of world is a persistent danger to public health. Currently there is no single treatment to eradicate the risk of COVID-19. The widespread transmission of SARS-CoV-2 due to the Omicron variant necessitates continued work on the development and implementation of effective vaccines. Moreover, there is evidence that mutations in the receptor domain of the SARS-CoV-2 spike glycoprotein led to the decrease in current vaccine efficacy by escaping antibody recognition. Therefore, it is essential to actively identify the mechanisms by which SARS-CoV-2 evades the host immune system, study the long-lasting effects of COVID-19 and develop therapeutics targeting SARS-CoV-2 infections in humans and preclinical models. In this review, we describe the pathogenic mechanisms of SARS-CoV-2 infection as well as the innate and adaptive host immune responses to infection. We address the ongoing need to develop effective vaccines that provide protection against different variants of SARS-CoV-2, as well as validated endpoint assays to evaluate the immunogenicity of vaccines in the pipeline, medications, anti-viral drug therapies and public health measures, that will be required to successfully end the COVID-19 pandemic.
Collapse
Affiliation(s)
- Rakhi Harne
- Seattle Children's Research Institute, Center for Global Infectious Disease Research, Seattle Children's Hospital, Seattle, Washington, USA
| | - Brittany Williams
- Seattle Children's Research Institute, Center for Global Infectious Disease Research, Seattle Children's Hospital, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Hazem F. M. Abdelaal
- Seattle Children's Research Institute, Center for Global Infectious Disease Research, Seattle Children's Hospital, Seattle, Washington, USA
| | - Susan L. Baldwin
- Seattle Children's Research Institute, Center for Global Infectious Disease Research, Seattle Children's Hospital, Seattle, Washington, USA
| | - Rhea N. Coler
- Seattle Children's Research Institute, Center for Global Infectious Disease Research, Seattle Children's Hospital, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
28
|
Regev-Yochay G, Lustig Y, Joseph G, Gilboa M, Barda N, Gens I, Indenbaum V, Halpern O, Katz-Likvornik S, Levin T, Kanaaneh Y, Asraf K, Amit S, Rubin C, Ziv A, Koren R, Mandelboim M, Tokayer NH, Meltzer L, Doolman R, Mendelson E, Alroy-Preis S, Kreiss Y. Correlates of protection against COVID-19 infection and intensity of symptomatic disease in vaccinated individuals exposed to SARS-CoV-2 in households in Israel (ICoFS): a prospective cohort study. THE LANCET. MICROBE 2023; 4:e309-e318. [PMID: 36963419 PMCID: PMC10030121 DOI: 10.1016/s2666-5247(23)00012-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 01/11/2023] [Accepted: 01/19/2023] [Indexed: 03/24/2023]
Abstract
BACKGROUND Identifying COVID-19 correlates of protection and immunity thresholds is important for policy makers and vaccine development. We aimed to identify correlates of protection of BNT162b2 (Pfizer-BioNTech) vaccination against COVID-19. METHODS In this prospective cohort study, households within a radius of 40 km of the Sheba Medical Center in Israel in which a new SARS-CoV-2 infection (defined as the index case) was detected within the previous 24 h were approached between July 25 and Nov 15, 2021. We included adults (aged >18 years) who had received one or two vaccine doses, had an initial negative SARS-CoV-2 PCR and no previous infection reported, and had a valid IgG and neutralising antibody result. The exposure of interest was baseline immune status, including IgG antibody concentration, neutralising antibody titre, and T-cell activation. The outcomes of interest were PCR-positive SARS-CoV-2 infection between day 2 and day 21 of follow-up and intensity of disease symptoms (self-reported via a telephone questionnaire) among participants who had a confirmed infection. Multivariable logistic and ordered logit ordinal regressions were used for the adjusted analysis. To identify immunological thresholds for clinical protection, we estimated the conditional probability of infection and moderate or severe disease for individuals with pre-exposure IgG and neutralising antibody concentrations above each value observed in the study data. FINDINGS From 16 675 detected index cases in the study region, 5718 household members agreed to participate, 1461 of whom were eligible to be included in our study. 333 (22·8%) of 1461 household members who were not infected with SARS-CoV-2 at baseline were infected within 21 days of follow-up. The baseline (pre-exposure) IgG and neutralising antibodies were higher in participants who remained uninfected than in those who became infected (geometric mean IgG antibody concentration 168·2 binding antibody units [BAU] per mL [95% CI 158·3-178·7] vs 130·5 BAU/mL [118·3-143·8] and geometric mean neutralising antibody titre 197·5 [181·9-214·4] vs 136 ·7 [120·3-155·4]). Increasing IgG and neutralising antibody concentrations were also significantly associated with a reduced probability of increasing disease severity. Odds of infection were significantly reduced each time baseline IgG antibody concentration increased by a factor of ten (odds ratio [OR] 0·43 [95% CI 0·26-0·70]) and each time baseline neutralising antibody titre increased by a factor of two (0·82 [0·74-0·92]). In our cohort, the probability of infection if IgG antibody concentrations were higher than 500 BAU/mL was 11% and the probability of moderate disease severity was 1%; the probability of infection if neutralising antibody titres were above or equal to 1024 was 8% and the probability of moderate disease severity was 2%. T-cell activation rates were not significantly associated with reduced probability of infection (OR 1·04, 95% CI 0·83-1·30). INTERPRETATION Both IgG and neutralising antibodies are correlates of protection against SARS-CoV-2 infection. Our data suggest that IgG concentrations higher than 500 BAU/mL and neutralising antibody titres of 1024 or more are thresholds for immunological protection from SARS-CoV-2 delta variant infection. Potentially, updated protective thresholds against emerging variants of concern could be calculated, which could support decision makers on administration of new vaccination strategies and on the optimal period between vaccine doses. FUNDING Israeli Ministry of Health.
Collapse
Affiliation(s)
- Gili Regev-Yochay
- Infection Prevention & Control Unit, Sheba Medical Center, Ramat Gan, Israel; Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel.
| | - Yaniv Lustig
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel; Central Virology Laboratory, Public Health Services, Ministry of Health, Ramat Gan, Israel
| | - Gili Joseph
- Infection Prevention & Control Unit, Sheba Medical Center, Ramat Gan, Israel
| | - Mayan Gilboa
- Infection Prevention & Control Unit, Sheba Medical Center, Ramat Gan, Israel; Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Noam Barda
- ARC Innovation Center, Sheba Medical Center, Ramat Gan, Israel; Department of Software and Information Systems Engineering, and Department of Epidemiology, Biostatistics and Community Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Ilana Gens
- Public Health Services, Israeli Ministry of Health, Jerusalem, Israel
| | - Victoria Indenbaum
- Central Virology Laboratory, Public Health Services, Ministry of Health, Ramat Gan, Israel
| | - Osnat Halpern
- Central Virology Laboratory, Public Health Services, Ministry of Health, Ramat Gan, Israel
| | - Shiri Katz-Likvornik
- Central Virology Laboratory, Public Health Services, Ministry of Health, Ramat Gan, Israel
| | - Tal Levin
- Central Virology Laboratory, Public Health Services, Ministry of Health, Ramat Gan, Israel
| | - Yara Kanaaneh
- Central Virology Laboratory, Public Health Services, Ministry of Health, Ramat Gan, Israel
| | - Keren Asraf
- The Dworman Automated-Mega Laboratory, Sheba Medical Center, Ramat Gan, Israel
| | - Sharon Amit
- Clinical Microbiology, Sheba Medical Center, Ramat Gan, Israel; Clinical Microbiology, Sheba Medical Center, Ramat Gan, Israel
| | - Carmit Rubin
- Infection Prevention & Control Unit, Sheba Medical Center, Ramat Gan, Israel
| | - Arnona Ziv
- Data Management Unit, Gerner Institute, Sheba Medical Center, Ramat Gan, Israel
| | - Ravit Koren
- Central Virology Laboratory, Public Health Services, Ministry of Health, Ramat Gan, Israel
| | - Michal Mandelboim
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel; Central Virology Laboratory, Public Health Services, Ministry of Health, Ramat Gan, Israel
| | - Noam H Tokayer
- Infection Prevention & Control Unit, Sheba Medical Center, Ramat Gan, Israel
| | - Lilac Meltzer
- Infection Prevention & Control Unit, Sheba Medical Center, Ramat Gan, Israel
| | - Ram Doolman
- Data Management Unit, Gerner Institute, Sheba Medical Center, Ramat Gan, Israel
| | - Ella Mendelson
- Central Virology Laboratory, Public Health Services, Ministry of Health, Ramat Gan, Israel
| | | | - Yitshak Kreiss
- General Management, Sheba Medical Center, Ramat Gan, Israel; Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| |
Collapse
|
29
|
Panagoulias I, Charokopos N, Thomas I, Spantidea PI, de Lastic AL, Rodi M, Anastasopoulou S, Aggeletopoulou I, Lazaris C, Karkoulias K, Leonidou L, Georgopoulos NA, Markou KB, Mouzaki A. Shifting gears: Study of immune system parameters of male habitual marathon runners. Front Immunol 2023; 13:1009065. [PMID: 36713459 PMCID: PMC9880332 DOI: 10.3389/fimmu.2022.1009065] [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: 08/01/2022] [Accepted: 12/27/2022] [Indexed: 01/15/2023] Open
Abstract
Aim Marathon is a running event in which athletes must cover a distance of 42.195 km. In addition to participating in marathons, marathoners have incorporated extensive running into their lifestyle. In the present study, we investigated the effect of long-term strenuous exercise in the form of marathon running on the immune system. Methods & Results We collected peripheral blood samples from 37 male marathoners before/after a race and 37 age/sex/body mass index (BMI)-matched healthy sedentary controls. Hematological and biochemical tests revealed race-induced leukocytosis attributable to neutrophilia and significant increases in plasma lactate dehydrogenase (LDH), creatine phosphokinase (CPK), and cortisol concentrations. Phenotypic analysis of lymphocytes revealed race-induced significant decrease in the number of lymphocytes, memory helper T (Th) cells, naive, memory and activated cytotoxic T (Tc) cells, natural killer (NK), NKT, and B1 cells, and a significant increase in the number of activated Th and regulatory Th cells (Tregs). Compared with controls, marathoners maintained significantly lower levels of memory and activated Th cells and higher levels of activated Tc and B1 cells. Measurement of plasma cytokine levels revealed a pro-inflammatory cytokine polarization that increased after the race. Examination of gene expression of cytokines and Th-cell signature transcription factors in peripheral blood mononuclear cells revealed a significant decrease in tumor necrosis factor α (TNF-α) and interleukin (IL)-17, and a significant increase in IL-6, IL-10 and forkhead box P3 (FoxP3) after the race. Compared with controls, marathoners maintained significantly higher levels of TNF-α. Assessment of the suppressive capacity of Tregs in co-cultures of isolated effector Th cells and Tregs showed significantly increased suppressive capacity of marathoners' Tregs after the race. Conclusions Compared with controls, marathoners live with permanent changes in certain immune parameters. Marathoners exhibit a stable pro-inflammatory cytokine polarization that increases after the race and is counterbalanced by increased numbers of Tregs overexpressing FoxP3 and having increased suppressive capacity.
Collapse
Affiliation(s)
- Ioannis Panagoulias
- Laboratory of Immunohematology, Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Nikolaos Charokopos
- Department of Respiratory Medicine, General Hospital of Pyrgos “Andreas Papandreou”, Pyrgos, Greece
| | - Iason Thomas
- Allergy Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Panagiota I. Spantidea
- Laboratory of Immunohematology, Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Anne-Lise de Lastic
- Laboratory of Immunohematology, Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Maria Rodi
- Laboratory of Immunohematology, Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Spyridoula Anastasopoulou
- Laboratory of Immunohematology, Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Ioanna Aggeletopoulou
- Laboratory of Immunohematology, Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Charalampos Lazaris
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Kiriakos Karkoulias
- Division of Respiratory Medicine, Department of Internal Medicine, Patras University Hospital, Patras, Greece
| | - Lydia Leonidou
- Division of Infectious Diseases, Department of Internal Medicine, Patras University Hospital, Patras, Greece
| | - Neoklis A. Georgopoulos
- Division of Endocrinology, Department of Internal Medicine, Patras University Hospital, Patras, Greece
| | - Kostas B. Markou
- Division of Endocrinology, Department of Internal Medicine, Patras University Hospital, Patras, Greece
| | - Athanasia Mouzaki
- Laboratory of Immunohematology, Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece,*Correspondence: Athanasia Mouzaki,
| |
Collapse
|
30
|
Kodali L, Budhiraja P, Gea-Banacloche J. COVID-19 in kidney transplantation-implications for immunosuppression and vaccination. Front Med (Lausanne) 2022; 9:1060265. [PMID: 36507509 PMCID: PMC9727141 DOI: 10.3389/fmed.2022.1060265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2022] Open
Abstract
COVID-19 pandemic continues to challenge the transplant community, given increased morbidity and mortality associated with the disease and poor response to prevention measures such as vaccination. Transplant recipients have a diminished response to both mRNA and vector-based vaccines compared to dialysis and the general population. The currently available assays to measure response to vaccination includes commercially available antibody assays for anti-Spike Ab, or anti- Receptor Binding Domain Ab. Positive antibody testing on the assays does not always correlate with neutralizing antibodies unless the antibody levels are high. Vaccinations help with boosting polyfunctional CD4+ T cell response, which continues to improve with subsequent booster doses. Ongoing efforts to improve vaccine response by using additional booster doses and heterologous vaccine combinations are underway. There is improved antibody response in moderate responders; however, the ones with poor response to initial vaccination doses, continue to have a poor response to sequential boosters. Factors associated with poor vaccine response include diabetes, older age, specific immunosuppressants such as belatacept, and high dose mycophenolate. In poor responders, a decrease in immunosuppression can increase response to vaccination. COVID infection or vaccination has not been associated with an increased risk of rejection. Pre- and Post-exposure monoclonal antibodies are available to provide further protection against COVID infection, especially in poor vaccine responders. However, the efficacy is challenged by the emergence of new viral strains. A recently approved bivalent vaccine offers better protection against the Omicron variant.
Collapse
Affiliation(s)
- Lavanya Kodali
- Department of Internal Medicine, Mayo Clinic, Phoenix, AZ, United States
- Division of Nephrology, Transplant Center, Mayo Clinic, Phoenix, AZ, United States
| | - Pooja Budhiraja
- Department of Internal Medicine, Mayo Clinic, Phoenix, AZ, United States
- Division of Nephrology, Transplant Center, Mayo Clinic, Phoenix, AZ, United States
| | - Juan Gea-Banacloche
- Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| |
Collapse
|
31
|
Silva MJA, Ribeiro LR, Lima KVB, Lima LNGC. Adaptive immunity to SARS-CoV-2 infection: A systematic review. Front Immunol 2022; 13:1001198. [PMID: 36300105 PMCID: PMC9589156 DOI: 10.3389/fimmu.2022.1001198] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 09/26/2022] [Indexed: 01/08/2023] Open
Abstract
Background There is evidence that the adaptive or acquired immune system is one of the crucial variables in differentiating the course of coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This work aimed to analyze the immunopathological aspects of adaptive immunity that are involved in the progression of this disease. Methods This is a systematic review based on articles that included experimental evidence from in vitro assays, cohort studies, reviews, cross-sectional and case-control studies from PubMed, SciELO, MEDLINE, and Lilacs databases in English, Portuguese, or Spanish between January 2020 and July 2022. Results Fifty-six articles were finalized for this review. CD4+ T cells were the most resolutive in the health-disease process compared with B cells and CD8+ T lymphocytes. The predominant subpopulations of T helper lymphocytes (Th) in critically ill patients are Th1, Th2, Th17 (without their main characteristics) and regulatory T cells (Treg), while in mild cases there is an influx of Th1, Th2, Th17 and follicular T helper cells (Tfh). These cells are responsible for the secretion of cytokines, including interleukin (IL) - 6, IL-4, IL-10, IL-7, IL-22, IL-21, IL-15, IL-1α, IL-23, IL-5, IL-13, IL-2, IL-17, tumor necrosis factor alpha (TNF-α), CXC motivating ligand (CXCL) 8, CXCL9 and tumor growth factor beta (TGF-β), with the abovementioned first 8 inflammatory mediators related to clinical benefits, while the others to a poor prognosis. Some CD8+ T lymphocyte markers are associated with the severity of the disease, such as human leukocyte antigen (HLA-DR) and programmed cell death protein 1 (PD-1). Among the antibodies produced by SARS-CoV-2, Immunoglobulin (Ig) A stood out due to its potent release associated with a more severe clinical form. Conclusions It is concluded that through this study it is possible to have a brief overview of the main immunological biomarkers and their function during SARS-CoV-2 infection in particular cell types. In critically ill individuals, adaptive immunity is varied, aberrantly compromised, and late. In particular, the T-cell response is also an essential and necessary component in immunological memory and therefore should be addressed in vaccine formulation strategies.
Collapse
Affiliation(s)
- Marcos Jessé Abrahão Silva
- Graduate Program in Epidemiology and Health Surveillance (PPGEVS), Bacteriology and Mycology Section (SABMI), Evandro Chagas Institute (IEC), Ananindeua, Brazil
- Bacteriology and Mycology Section (SABMI), Evandro Chagas Institute (IEC), Ananindeua, Brazil
- *Correspondence: Marcos Jessé Abrahão Silva,
| | - Layana Rufino Ribeiro
- Bacteriology and Mycology Section (SABMI), Evandro Chagas Institute (IEC), Ananindeua, Brazil
| | | | - Luana Nepomuceno Gondim Costa Lima
- Graduate Program in Epidemiology and Health Surveillance (PPGEVS), Bacteriology and Mycology Section (SABMI), Evandro Chagas Institute (IEC), Ananindeua, Brazil
- Bacteriology and Mycology Section (SABMI), Evandro Chagas Institute (IEC), Ananindeua, Brazil
| |
Collapse
|
32
|
Vaccine-Induced T-Cell and Antibody Responses at 12 Months after Full Vaccination Differ with Respect to Omicron Recognition. Vaccines (Basel) 2022; 10:vaccines10091563. [PMID: 36146641 PMCID: PMC9500953 DOI: 10.3390/vaccines10091563] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 12/22/2022] Open
Abstract
More than a year after the first vaccines against the novel SARS-CoV-2 were approved, many questions still remain about the long-term protection conferred by each vaccine. How long the effect lasts, how effective it is against variants of concern and whether further vaccinations will confer additional benefits remain part of current and future research. For this purpose, we examined 182 health care employees-some of them with previous SARS-CoV-2 infection-12 months after different primary immunizations. To assess antibody responses, we performed an electrochemiluminescence assay (ECLIA) to determine anti-spike IgGs, followed by a surrogate virus neutralization assay against Wuhan-Hu-1 and B.1.1.529/BA.1 (Omicron). T cell response against wild-type and the Omicron variants of concern were assessed via interferon-gamma ELISpot assays and T-cell surface and intracellular cytokine staining. In summary, our results show that after the third vaccination with an mRNA vaccine, differences in antibody quantity and functionality observed after different primary immunizations were equalized. As for the T cell response, we were able to demonstrate a memory function for CD4+ and CD8+ T cells alike. Importantly, both T and antibody responses against wild-type and omicron differed significantly; however, antibody and T cell responses did not correlate with each other and, thus, may contribute differentially to immunity.
Collapse
|