1
|
Meyers E, De Rop L, Gioveni C, Engels F, Coen A, De Burghgraeve T, Digregorio M, Van Ngoc P, De Clercq N, Buret L, Coenen S, Padalko E, Duysburgh E, Scholtes B, Verbakel JY, Heytens S, Cools P. Dynamics of SARS-CoV-2 IgG in Nursing Home Residents in Belgium Throughout Three BNT162b2 Vaccination Rounds: 19-Month Follow-Up. Vaccines (Basel) 2025; 13:409. [PMID: 40333301 PMCID: PMC12030799 DOI: 10.3390/vaccines13040409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/03/2025] [Accepted: 04/12/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND/OBJECTIVES This study mapped antibody dynamics across three COVID-19 vaccination rounds (primary course, first, and second booster with BNT162b2) in Belgian nursing home residents (NHRs). METHODS Within a national SARS-CoV-2 serosurveillance study (February 2021-September 2022) across Belgian nursing homes, dried blood spots were collected, on which anti-spike SARS-CoV-2 IgG antibodies were quantified by ELISA in international units/mL (IU/mL). Sociodemographic data were collected at the study start and infection history and vaccination data at each sampling round. RESULTS Infection-naïve NHRs had low antibody levels after primary course vaccination (geometric mean concentration (GMC) 292 IU/mL, 95% confidence interval (95% CI): 197-432), but increased tenfold after first booster (GMC 2168 IU/mL, 95% CI: 1554-3027). While antibodies among NHRs significantly declined within six months after primary vaccination (p < 0.0001), they remained stable for nine months post-booster (p > 0.05). Among primary vaccine non-responders, 92% (95% CI: 82-97%) developed antibodies after the first booster (GMC 594 IU/mL, 95% CI: 416-849), though tenfold lower than initial responders (GMC 4642 IU/mL, 95% CI: 3577-6022). CONCLUSIONS These findings demonstrate that NHRs require tailored vaccination, prioritizing repeated immunization to improve serological outcomes in poor responders such as infection-naive NHRs. Regular immune monitoring could aid in implementing evidence-based vaccine strategies, ensuring optimal protection for vulnerable populations against SARS-CoV-2 and other infectious threats.
Collapse
Affiliation(s)
- Eline Meyers
- Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; (E.M.)
| | - Liselore De Rop
- LUHTAR—Leuven Unit for HTA Research, Department of Public Health and Primary Care, KU Leuven, 3000 Leuven, Belgium; (L.D.R.); (T.D.B.); (J.Y.V.)
| | - Claudia Gioveni
- Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; (E.M.)
| | - Fien Engels
- Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; (E.M.)
| | - Anja Coen
- Department of Public Health and Primary Care, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Tine De Burghgraeve
- LUHTAR—Leuven Unit for HTA Research, Department of Public Health and Primary Care, KU Leuven, 3000 Leuven, Belgium; (L.D.R.); (T.D.B.); (J.Y.V.)
| | - Marina Digregorio
- Research Unit of Primary Care and Health, Department of General Medicine, Faculty of Medicine, University of Liège, 4000 Liège, Belgium (B.S.)
| | - Pauline Van Ngoc
- Research Unit of Primary Care and Health, Department of General Medicine, Faculty of Medicine, University of Liège, 4000 Liège, Belgium (B.S.)
| | - Nele De Clercq
- Department of Public Health and Primary Care, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Laëtitia Buret
- Research Unit of Primary Care and Health, Department of General Medicine, Faculty of Medicine, University of Liège, 4000 Liège, Belgium (B.S.)
| | - Samuel Coenen
- Department of Family Medicine & Population Health, Faculty of Medicine and Health Sciences, University of Antwerp, 2000 Antwerp, Belgium;
| | - Elizaveta Padalko
- Laboratory of Medical Microbiology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Els Duysburgh
- Department of Epidemiology and Public Health, Sciensano, 1000 Brussels, Belgium
| | - Beatrice Scholtes
- Research Unit of Primary Care and Health, Department of General Medicine, Faculty of Medicine, University of Liège, 4000 Liège, Belgium (B.S.)
| | - Jan Y. Verbakel
- LUHTAR—Leuven Unit for HTA Research, Department of Public Health and Primary Care, KU Leuven, 3000 Leuven, Belgium; (L.D.R.); (T.D.B.); (J.Y.V.)
| | - Stefan Heytens
- Department of Public Health and Primary Care, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Piet Cools
- Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; (E.M.)
| |
Collapse
|
2
|
Fialova M, Cecrdlova E, Zahradka I, Petr V, Hruby F, Modos I, Viklicky O, Striz I. Attenuated neutralization, maintained specificity: Humoral response to SARS-CoV-2 booster in kidney allograft recipients. Diagn Microbiol Infect Dis 2025; 111:116550. [PMID: 39437653 DOI: 10.1016/j.diagmicrobio.2024.116550] [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: 07/08/2024] [Revised: 09/20/2024] [Accepted: 10/04/2024] [Indexed: 10/25/2024]
Abstract
Despite the lower virulence of current SARS-CoV-2 variants and high rates of vaccinated and previously infected subjects, COVID-19 remains a persistent threat in kidney transplant recipients (KTRs). This study evaluated the parameters of anti-SARS-CoV-2 antibody production in 120 KTRs. The production of neutralizing antibodies in KTRs, following booster vaccination with the mRNA vaccine BNT162b2, was significantly decreased and their decline was faster than in healthy subjects. Factors predisposing to the downregulation of anti-SARS-CoV-2 neutralizing antibodies included age, lower estimated glomerular filtration rate, and a full dose of mycophenolate mofetil. Neutralizing antibodies correlated with those targeting the SARS-CoV-2 receptor binding domain (RBD), SARS-CoV-2 Spike trimmer, total SARS-CoV-2 S1 protein, as well as with antibodies to the deadly SARS-CoV-1 virus. No cross-reactivity was found with antibodies against seasonal coronaviruses. KTRs exhibited lower postvaccination production of neutralizing antibodies against SARS-CoV-2; however, the specificity of their humoral response did not differ compared to healthy subjects.
Collapse
Affiliation(s)
- Martina Fialova
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Eva Cecrdlova
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Ivan Zahradka
- Department of Nephrology, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Vojtech Petr
- Department of Nephrology, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Filip Hruby
- Information Technology Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Istvan Modos
- Information Technology Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Ondrej Viklicky
- Department of Nephrology, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Ilja Striz
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| |
Collapse
|
3
|
Infante-Domínguez C, Salto-Alejandre S, Álvarez-Marín R, Sabé N, Ramos-Martínez A, Moreno A, Ferreira de Moraes K, Palacios-Baena ZR, Muñoz P, Fernández-Ruiz M, Blanes M, Fariñas C, Vidal E, Merino de Lucas E, Halpern M, Hernández-Gallego R, Bassetti M, Mularoni A, Gutiérrez-Dalmau A, Rinaldi M, Jiménez-Jorge S, Bodro M, Aranha-Camargo LF, Valerio M, Sánchez-Céspedes J, Gutiérrez-Gutiérrez B, Giannella M, Rodríguez-Baño J, Pachón J, Cordero E. COVID-19 clinical phenotypes in vaccinated and nonvaccinated solid organ transplant recipients: a multicenter validation study. Sci Rep 2024; 14:30021. [PMID: 39622951 PMCID: PMC11612230 DOI: 10.1038/s41598-024-81099-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 11/25/2024] [Indexed: 12/06/2024] Open
Abstract
Clinical phenotypes of COVID-19, associated with mortality risk, have been identified in the general population. The present study assesses their applicability in solid organ transplant recipients (SOTR) hospital-admitted by COVID-19. In a cohort of 488 SOTR, nonvaccinated (n = 394) and vaccinated (n = 94) against SARS-CoV-2, we evaluated 16 demographic, clinical, analytical, and radiological variables to identify the clinical phenotypes A, B, and C. The median age was 61.0 (51-69) years, 330 (67.6%) and 158 (32.4%) were men and women, respectively, 415 (85%) had pneumonia, and 161 (33%) had SpO2 < 95% at admission. All-cause mortality occurred in 105 (21.5%) cases. It was higher in nonvaccinated versus vaccinated SOTR (23.4% vs 13.8%, P = 0.04). Patients in the entire cohort were classified into phenotypes A (n = 149, 30.5%), B (n = 187, 38.3%), and C (n = 152, 31.1%), with mortality rates of 8.7%, 16.6%, and 40.1%, respectively, which were similar to those of nonvaccinated SOTR (9.5%, 16.7%, and 52.0%) and lower in vaccinated SOTR (4.4%, 15.8%, and 17.3%, respectively), with difference between nonvaccinated and vaccinated in the phenotype C (P < 0.001). In conclusion, COVID-19 clinical phenotypes are useful in SOTR, and all-cause mortality decreases in vaccinated patients.
Collapse
Affiliation(s)
- Carmen Infante-Domínguez
- Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Instituto de Biomedicina de Sevilla (IBiS), Virgen del Rocío University Hospital /CSIC/University of Seville, Seville, Spain
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Sonsoles Salto-Alejandre
- Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Instituto de Biomedicina de Sevilla (IBiS), Virgen del Rocío University Hospital /CSIC/University of Seville, Seville, Spain
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Rocío Álvarez-Marín
- Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Instituto de Biomedicina de Sevilla (IBiS), Virgen del Rocío University Hospital /CSIC/University of Seville, Seville, Spain
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Nuria Sabé
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Service of Infectious Diseases, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), University of Barcelona, Hospitalet de Llobregat, Barcelona, Spain
| | - Antonio Ramos-Martínez
- Unit of Infectious Diseases, Microbiology, and Preventive Medicine. Hospital, Universitario Puerta de Hierro-Majadahonda-IDIPHISA, Madrid, Spain
| | - Asunción Moreno
- Service of Infectious Diseases, Hospital Clinic-IDIBAPS. University of Barcelona, Barcelona, Spain
| | | | - Zaira R Palacios-Baena
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Unit of Infectious Diseases and Microbiology, Virgen Macarena University Hospital, Instituto de Biomedicina de Sevilla (IBiS)/CSIC/University of Seville, Seville, Spain
| | - Patricia Muñoz
- Clinical Microbiology and Infectious Diseases Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CIBERES, ISCIII-CIBER de Enfermedades Respiratorias. Medicine Department, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Mario Fernández-Ruiz
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Unit of Infectious Diseases, "12 de Octubre" University Hospital, Research Institute Hospital "12 de Octubre" (I+12), Madrid, Spain
| | - Marino Blanes
- Unit of Infectious Diseases, La Fe University Hospital, Valencia, Spain
| | - Carmen Fariñas
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Service of Infectious Diseases, Marqués de Valdecilla University Hospital, Marqués de Valdecilla-IDIVAL, University of Cantabria, Santander, Spain
| | - Elisa Vidal
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Service of Infectious Diseases, Reina Sofía University Hospital, Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
| | - Esperanza Merino de Lucas
- Unit of Infectious Diseases, Alicante General University Hospital, Alicante Institute of Health and Biomedical Research (ISABIAL), Alicante, Spain
- Clinical Medicine Department, Miguel Hernández University, Elche, Spain
| | - Márcia Halpern
- Liver Transplantation Program, Quinta D'Or Hospital, Rio de Janeiro, Brazil
| | - Román Hernández-Gallego
- Unit of Kidney Transplant, Service of Nephrology, Badajoz University Hospital, Badajoz, Spain
| | - Matteo Bassetti
- Infectious Diseases Clinic, Policlinico San Martino Hospital-IRCCS, Department of Health Science, University of Genoa, Genoa, Italy
| | - Alessandra Mularoni
- Unit of Infectious Diseases and Infection Control, ISMETT-IRCCS-Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione-Istituto di Ricovero e Cura a Carattere Scientifico, Palermo, Italy
| | - Alex Gutiérrez-Dalmau
- Renal Transplant Unit, Nephrology Service, Miguel Servet University Hospital, Aragón Institute for Health Research IIS-Aragón, Zaragoza, Spain
| | - Matteo Rinaldi
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
- Infectious Diseases Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - Silvia Jiménez-Jorge
- Clinical Research and Clinical Trials Unit (CTU), Instituto de Biomedicina de Sevilla (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Seville, Spain
| | - Marta Bodro
- Service of Infectious Diseases, Hospital Clinic-IDIBAPS. University of Barcelona, Barcelona, Spain
| | | | - Maricela Valerio
- Clinical Microbiology and Infectious Diseases Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CIBERES, ISCIII-CIBER de Enfermedades Respiratorias. Medicine Department, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Javier Sánchez-Céspedes
- Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Instituto de Biomedicina de Sevilla (IBiS), Virgen del Rocío University Hospital /CSIC/University of Seville, Seville, Spain
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Belén Gutiérrez-Gutiérrez
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Unit of Infectious Diseases and Microbiology, Virgen Macarena University Hospital, Instituto de Biomedicina de Sevilla (IBiS)/CSIC/University of Seville, Seville, Spain
- Department of Medicine, School of Medicine, University of Seville, Seville, Spain
| | - Maddalena Giannella
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
- Infectious Diseases Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - Jesús Rodríguez-Baño
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Unit of Infectious Diseases and Microbiology, Virgen Macarena University Hospital, Instituto de Biomedicina de Sevilla (IBiS)/CSIC/University of Seville, Seville, Spain
- Department of Medicine, School of Medicine, University of Seville, Seville, Spain
| | - Jerónimo Pachón
- Department of Medicine, School of Medicine, University of Seville, Seville, Spain.
- Instituto de Biomedicina de Sevilla (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Av. Manuel Siurot s/n, 41013, Seville, Spain.
| | - Elisa Cordero
- Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Instituto de Biomedicina de Sevilla (IBiS), Virgen del Rocío University Hospital /CSIC/University of Seville, Seville, Spain
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Department of Medicine, School of Medicine, University of Seville, Seville, Spain
| |
Collapse
|
4
|
Favà A, Couceiro C, Calatayud L, Hernandez-Hermida Y, Melilli E, Montero N, Manonelles A, Coloma A, Codina S, Lloberas N, Oliveras L, Lino LA, Galofré C, Sabé N, Gomez-Preciado F, Sandoval D, Pizarro D, Domínguez MA, Cruzado JM. Hybrid immunity protection against SARS-CoV-2 and severe COVID-19 in kidney transplantation: A retrospective, comparative cohort study. Am J Transplant 2024; 24:2282-2291. [PMID: 39097095 DOI: 10.1016/j.ajt.2024.07.028] [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: 03/27/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/05/2024]
Abstract
Hybrid immunity, resulting from a combination of SARS-CoV-2 infection and vaccination, offers robust protection against COVID-19 in the general population. However, its impact on immunocompromised patients remains unexplored. We investigated the effect of hybrid immunity against the Omicron variant in a population of kidney transplant recipients receiving the fourth dose mRNA monovalent vaccination. By extracting data from the clinical records and performing individual interviews, participants were categorized into the hybrid cohort (previously infected and vaccinated individuals) and the vaccine cohort (vaccinated-only individuals). The study comprised 1114 participants, 442 in the hybrid and 672 in the vaccine cohorts. From April 2022 to August 2023, 286 infections, 38 hospitalizations and 9 deaths were reported. The cumulative incidence of infection was 12.1% (95% confidence interval [CI], 9.03-16.03) for the hybrid cohort and 36.54% (95% CI, 32.81-40.54) for the vaccine cohort after 300 days of follow-up. Hybrid immunity was associated to a 72% lower risk of infection (adjusted hazard ratio, 0.28; 95% CI, 0.21-0.38) and a 96% lower risk of hospitalization (adjusted hazard ratio, 0.04; 95% CI, 0.01-0.32). No deaths occurred in the hybrid cohort. Hybrid immunity was associated with a lower incidence of SARS-CoV-2 infection and severe COVID-19, underscoring its importance for risk stratification in this vulnerable patient population.
Collapse
Affiliation(s)
- Alexandre Favà
- Nephrology Department, Bellvitge University Hospital, Barcelona, Spain; Nephrology and Renal Transplantation Group, Infectious Disease and Transplantation Program, Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, Spain.
| | - Carlos Couceiro
- Nephrology Department, Bellvitge University Hospital, Barcelona, Spain; Nephrology and Renal Transplantation Group, Infectious Disease and Transplantation Program, Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, Spain.
| | - Laura Calatayud
- Microbiology Department, Bellvitge University Hospital, Barcelona, Spain; Research Network for Respiratory Diseases (CIBERES), ISCIII, Madrid, Spain
| | | | - Edoardo Melilli
- Nephrology Department, Bellvitge University Hospital, Barcelona, Spain; Nephrology and Renal Transplantation Group, Infectious Disease and Transplantation Program, Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, Spain
| | - Nuria Montero
- Nephrology Department, Bellvitge University Hospital, Barcelona, Spain; Nephrology and Renal Transplantation Group, Infectious Disease and Transplantation Program, Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, Spain
| | - Anna Manonelles
- Nephrology Department, Bellvitge University Hospital, Barcelona, Spain; Nephrology and Renal Transplantation Group, Infectious Disease and Transplantation Program, Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, Spain; Department of Clinical Sciences, University of Barcelona, Barcelona, Spain
| | - Ana Coloma
- Nephrology Department, Bellvitge University Hospital, Barcelona, Spain; Nephrology and Renal Transplantation Group, Infectious Disease and Transplantation Program, Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, Spain
| | - Sergi Codina
- Nephrology Department, Bellvitge University Hospital, Barcelona, Spain; Nephrology and Renal Transplantation Group, Infectious Disease and Transplantation Program, Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, Spain
| | - Nuria Lloberas
- Nephrology Department, Bellvitge University Hospital, Barcelona, Spain; Nephrology and Renal Transplantation Group, Infectious Disease and Transplantation Program, Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, Spain; Department of Clinical Sciences, University of Barcelona, Barcelona, Spain
| | - Laia Oliveras
- Nephrology Department, Bellvitge University Hospital, Barcelona, Spain; Nephrology and Renal Transplantation Group, Infectious Disease and Transplantation Program, Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, Spain
| | - Luis Arturo Lino
- Nephrology Department, Bellvitge University Hospital, Barcelona, Spain
| | - Claudia Galofré
- Nephrology Department, Bellvitge University Hospital, Barcelona, Spain; Nephrology and Renal Transplantation Group, Infectious Disease and Transplantation Program, Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, Spain
| | - Nuria Sabé
- Department of Infectious Diseases, Bellvitge University Hospital, Barcelona, Spain
| | - Francisco Gomez-Preciado
- Nephrology Department, Bellvitge University Hospital, Barcelona, Spain; Nephrology and Renal Transplantation Group, Infectious Disease and Transplantation Program, Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, Spain
| | - Diego Sandoval
- Nephrology Department, Bellvitge University Hospital, Barcelona, Spain
| | - Daniel Pizarro
- Microbiology Department, Bellvitge University Hospital, Barcelona, Spain
| | - Maria Angeles Domínguez
- Microbiology Department, Bellvitge University Hospital, Barcelona, Spain; Research Network for Infectious Diseases (CIBERINFEC), ISCIII, Madrid, Spain; Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain
| | - Josep Maria Cruzado
- Nephrology Department, Bellvitge University Hospital, Barcelona, Spain; Nephrology and Renal Transplantation Group, Infectious Disease and Transplantation Program, Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, Spain; Department of Clinical Sciences, University of Barcelona, Barcelona, Spain
| |
Collapse
|
5
|
Augello M, Wagenhäuser I, Krone M, Dauby N, Ferrara P, Sabbatucci M, Ruta S, Rezahosseini O, Velikov P, Gkrania-Klotsas E, Montes J, Franco-Paredes C, Goodman AL, Küçükkaya S, Tuells J, Harboe ZB, Epaulard O. Should SARS-CoV-2 serological testing be used in the decision to deliver a COVID-19 vaccine booster? A pro-con assessment. Vaccine 2024; 42:126184. [PMID: 39097440 DOI: 10.1016/j.vaccine.2024.126184] [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: 02/15/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/05/2024]
Abstract
Anti-SARS-CoV-2 vaccination has saved millions of lives in the past few years. To maintain a high level of protection, particularly in at-risk populations, booster doses are recommended to counter the waning of circulating antibody levels over time and the continuous emergence of immune escape variants of concern (VOCs). As anti-spike serology is now widely available, it may be considered a useful tool to identify individuals needing an additional vaccine dose, i.e., to screen certain populations to identify those whose plasma antibody levels are too low to provide protection. However, no recommendations are currently available on this topic. We reviewed the relevant supporting and opposing arguments, including areas of uncertainty, and concluded that in most populations, spike serology should not be used to decide about the administration of a booster dose. The main counterarguments are as follows: correlates of protection are imperfectly characterised, essentially owing to the emergence of VOCs; spike serology has an intrinsic inability to comprehensively reflect the whole immune memory; and booster vaccines are now VOC-adapted, while the commonly available commercial serological assays explore antibodies against the original virus.
Collapse
Affiliation(s)
- Matteo Augello
- Clinic of Infectious Diseases and Tropical Medicine, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Isabell Wagenhäuser
- University Hospital Würzburg, Infection Control and Antimicrobial Stewardship Unit, Würzburg, Germany
| | - Manuel Krone
- University Hospital Würzburg, Infection Control and Antimicrobial Stewardship Unit, Würzburg, Germany
| | - Nicolas Dauby
- Department of Infectious Diseases, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Environmental health and occupational health, School of Public Health, Université Libre de Bruxelles (ULB), Brussel, Belgium
| | - Pietro Ferrara
- Center for Public Health Research, University of Milan - Bicocca, Monza, Italy; IRCCS Istituto Auxologico Italiano, Laboratory of Public Health, Milan, Italy
| | | | - Simona Ruta
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Stefan S Nicolau Institute of Virology, Bucharest, Romania
| | - Omid Rezahosseini
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital, Hillerød, Denmark
| | - Petar Velikov
- Clinic for Pediatric Infectious Diseases, Infectious Disease Hospital "Prof. Ivan Kirov", Sofia, Bulgaria; Department of Global Public Health, University of Tsukuba, Tsukuba, Japan
| | | | - Jose Montes
- Investigación en Resistencia Antibiótica (INVERA), Buenos Aires, Argentina; Fundación del Centro de Estudios Infectológicos (FUNCEI), Buenos Aires, Argentina
| | - Carlos Franco-Paredes
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, USA; Hospital Infantil de Mexico, Mexico City, Mexico
| | - Anna L Goodman
- Centre for Infection Diagnostics research, Department of Infection at at King's College London and Guys' and St Thomas NHS Foundation trust, London, UK
| | - Sertaç Küçükkaya
- Department of Medical Microbiology, Istanbul Faculty of Medicine, İstanbul University, Istanbul, Turkey
| | - Jose Tuells
- Departamento de Enfermería Comunitaria, Medicina Preventiva y Salud Pública e historia de la ciencia, Universidad de Alicante, Alicante, Spain
| | | | - Olivier Epaulard
- Université Grenoble Alpes, Infectiologie, CHU Grenoble Alpes, Grenoble, France.
| |
Collapse
|
6
|
Etienne I, Kemlin D, Gemander N, Olislagers V, Waegemans A, Dhondt E, Heyndrickx L, Depickère S, Charles A, Goossens M, Vandermosten L, Desombere I, Ariën KK, Pannus P, Knoop C, Marchant A. Persistent defect in SARS-CoV-2 humoral and cellular immunity in lung transplant recipients. J Heart Lung Transplant 2024; 43:1857-1860. [PMID: 39134165 DOI: 10.1016/j.healun.2024.08.002] [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: 03/06/2024] [Revised: 07/26/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024] Open
Abstract
Lung transplant recipients (LTRs) are susceptible to severe Coronavirus Disease 2019 (COVID-19) and had lower immune responses to primary severe acute respiratory syndrome-related to coronavirus 2 (SARS-CoV-2) vaccination as compared to the general population and to other solid organ transplant recipients. As immunity induced by booster vaccination and natural infection has increased since the beginning of the pandemic in the general population, immunity acquired by LTRs is not well documented. Humoral and cellular immunity to SARS-CoV-2 was monitored in February and May 2023 in 30 LTRs and compared to that of health care workers (HCWs) and nursing home residents (NHRs). LTRs had significantly lower levels of SARS-CoV-2 binding and neutralizing antibodies and lower interferon-gamma responses to Wuhan, Delta, and XBB1.5 variants as compared to HCWs and NHRs. Humoral immunity decreased between the 2 visits, whereas cellular immunity remained more stable. The persistent defect in SARS-CoV-2 immunity in LTRs should encourage continued monitoring and preventive measures for this vulnerable population.
Collapse
Affiliation(s)
- Isabelle Etienne
- Chest Department, Hôpital Universitaire de Bruxelles Erasme, Université Libre de Bruxelles, Brussels, Belgium; European Plotkin Institute for Vaccinology, Université Libre de Bruxelles, Brussels, Belgium.
| | - Delphine Kemlin
- European Plotkin Institute for Vaccinology, Université Libre de Bruxelles, Brussels, Belgium; Department of Nephrology, Dialysis and Transplantation, Hôpital Universitaire de Bruxelles Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Nicolas Gemander
- European Plotkin Institute for Vaccinology, Université Libre de Bruxelles, Brussels, Belgium; Department of Nephrology, Dialysis and Transplantation, Hôpital Universitaire de Bruxelles Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Véronique Olislagers
- European Plotkin Institute for Vaccinology, Université Libre de Bruxelles, Brussels, Belgium
| | - Alexandra Waegemans
- European Plotkin Institute for Vaccinology, Université Libre de Bruxelles, Brussels, Belgium
| | - Emilie Dhondt
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Leo Heyndrickx
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Stéphanie Depickère
- Plateform Interventional Studies, Scientific Direction Infectious Diseases in Humans, Sciensano, Brussels, Belgium
| | - Alexia Charles
- Plateform Interventional Studies, Scientific Direction Infectious Diseases in Humans, Sciensano, Brussels, Belgium
| | - Maria Goossens
- Plateform Interventional Studies, Scientific Direction Infectious Diseases in Humans, Sciensano, Brussels, Belgium
| | - Leen Vandermosten
- Laboratory of Immune Response, Scientific Direction Infectious Diseases in Humans, Sciensano, Brussels, Belgium
| | - Isabelle Desombere
- Laboratory of Immune Response, Scientific Direction Infectious Diseases in Humans, Sciensano, Brussels, Belgium
| | - Kevin K Ariën
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Pieter Pannus
- European Plotkin Institute for Vaccinology, Université Libre de Bruxelles, Brussels, Belgium
| | - Christiane Knoop
- Chest Department, Hôpital Universitaire de Bruxelles Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Arnaud Marchant
- European Plotkin Institute for Vaccinology, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
7
|
Espi M, Charmetant X, Benotmane I, Lefsihane K, Barateau V, Gallais F, Boulenouar H, Ovize A, Barbry A, Bouz C, Morelon E, Defrance T, Fafi-Kremer S, Caillard S, Thaunat O. Memory B Cells Provide Long-Term Protection to Vaccinated Kidney Transplant Recipients Against SARS-CoV-2 Variants. J Med Virol 2024; 96:e70037. [PMID: 39530340 DOI: 10.1002/jmv.70037] [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: 05/22/2024] [Revised: 09/10/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
Kidney transplant recipients (KTRs) are highly vulnerable to COVID-19. An intensified scheme of vaccination offers short-term protection to the 50%-75% of KTRs able to develop a germinal center reaction, required for the generation of neutralizing titers of antibodies (NAbs). However, the duration of this vaccinal protection is unknown. In-depth longitudinal analysis of the immune response to vaccination of 33 KTRs demonstrates that the low peak of IgGs, the progressive decline in antibody titers, and the emergence of a variant of concerns (VOC) of SARS-CoV2, synergize to let 2/3 of responders to vaccine without NAbs after only a few months. Yet, a retrospective study of an independent cohort of 274 KTRs, revealed that the risk of severe COVID-19 in the latter was low, similar to that of patients with serum neutralizing capacity against VOC. Our work links this late vaccine protection with the presence of memory B cells, which are generated during the initial vaccine-induced germinal center reaction, have a wide repertoire directed against conserved spike epitopes, and rapidly differentiate into IgG-producing plasma cells upon antigenic rechallenge. We conclude that in contrast with a serological layer that goes fading rapidly, the cellular layer of humoral memory provides an efficient long-term protection against VOC to KTRs. This illustration of the complementary roles of the two layers of the humoral memory has implications in immunopathology beyond the COVID-19 in KTRs.
Collapse
Affiliation(s)
- Maxime Espi
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Xavier Charmetant
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Lyon, France
- Department of Transplantation, Hospices Civils de Lyon, Edouard Herriot Hospital, Nephrology and Clinical Immunology, Lyon, France
- Claude Bernard University, Villeurbanne, France
| | - Ilies Benotmane
- Department of Nephrology Dialysis and Transplantation, Strasbourg University Hospital, Strasbourg, France
- Inserm UMR S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Katia Lefsihane
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Véronique Barateau
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Floriane Gallais
- Department of Virology, Strasbourg University Hospital, Strasbourg, France
| | - Hafsa Boulenouar
- Inserm UMR S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Anne Ovize
- Eurofins Biomnis Laboratory, Lyon, France
| | | | | | - Emmanuel Morelon
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Lyon, France
- Department of Transplantation, Hospices Civils de Lyon, Edouard Herriot Hospital, Nephrology and Clinical Immunology, Lyon, France
- Claude Bernard University, Villeurbanne, France
| | - Thierry Defrance
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Samira Fafi-Kremer
- Inserm UMR S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
- Department of Virology, Strasbourg University Hospital, Strasbourg, France
| | - Sophie Caillard
- Department of Nephrology Dialysis and Transplantation, Strasbourg University Hospital, Strasbourg, France
- Inserm UMR S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Olivier Thaunat
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Lyon, France
- Department of Transplantation, Hospices Civils de Lyon, Edouard Herriot Hospital, Nephrology and Clinical Immunology, Lyon, France
- Claude Bernard University, Villeurbanne, France
| |
Collapse
|
8
|
Schwalbe H, Audergon P, Haley N, Amaro CA, Agirre J, Baldus M, Banci L, Baumeister W, Blackledge M, Carazo JM, Carugo KD, Celie P, Felli I, Hart DJ, Hauß T, Lehtiö L, Lindorff-Larsen K, Márquez J, Matagne A, Pierattelli R, Rosato A, Sobott F, Sreeramulu S, Steyaert J, Sussman JL, Trantirek L, Weiss MS, Wilmanns M. The future of integrated structural biology. Structure 2024; 32:1563-1580. [PMID: 39293444 DOI: 10.1016/j.str.2024.08.014] [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: 05/23/2024] [Revised: 07/21/2024] [Accepted: 08/22/2024] [Indexed: 09/20/2024]
Abstract
Instruct-ERIC, "the European Research Infrastructure Consortium for Structural biology research," is a pan-European distributed research infrastructure making high-end technologies and methods in structural biology available to users. Here, we describe the current state-of-the-art of integrated structural biology and discuss potential future scientific developments as an impulse for the scientific community, many of which are located in Europe and are associated with Instruct. We reflect on where to focus scientific and technological initiatives within the distributed Instruct research infrastructure. This review does not intend to make recommendations on funding requirements or initiatives directly, neither at the national nor the European level. However, it addresses future challenges and opportunities for the field, and foresees the need for a stronger coordination within the European and international research field of integrated structural biology to be able to respond timely to thematic topics that are often prioritized by calls for funding addressing societal needs.
Collapse
Affiliation(s)
- Harald Schwalbe
- Center for Biomolecular Magnetic Resonance (BMRZ), Institute for Organic Chemistry, Max-von-Laue-Str. 7, 60438 Frankfurt/M., Germany; Instruct-ERIC, Oxford House, Parkway Court, John Smith Drive, Oxford OX4 2JY, UK.
| | - Pauline Audergon
- Instruct-ERIC, Oxford House, Parkway Court, John Smith Drive, Oxford OX4 2JY, UK
| | - Natalie Haley
- Instruct-ERIC, Oxford House, Parkway Court, John Smith Drive, Oxford OX4 2JY, UK
| | - Claudia Alen Amaro
- Instruct-ERIC, Oxford House, Parkway Court, John Smith Drive, Oxford OX4 2JY, UK
| | - Jon Agirre
- York Structural Biology Laboratory, Department of Chemistry, University of York, York YO10 3BG, UK
| | - Marc Baldus
- NMR Spectroscopy, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, Utrecht 3584 CH, the Netherlands
| | - Lucia Banci
- Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine-CIRMMP, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, Italy
| | - Wolfgang Baumeister
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Martin Blackledge
- Institut de Biologie Structurale, Université Grenoble Alpes-CEA-CNRS UMR5075, 71 Avenue des Martyrs, 38000 Grenoble, France
| | - Jose Maria Carazo
- Biocomputing Unit, National Centre for Biotechnology (CNB CSIC), Campus Universidad Autónoma de Madrid, Darwin 3, Cantoblanco, 28049 Madrid, Spain
| | | | - Patrick Celie
- Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Isabella Felli
- Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine-CIRMMP, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, Italy
| | - Darren J Hart
- Institut de Biologie Structurale, Université Grenoble Alpes-CEA-CNRS UMR5075, 71 Avenue des Martyrs, 38000 Grenoble, France
| | - Thomas Hauß
- Macromolecular Crystallography, Helmholtz-Zentrum, Albert-Einstein-Str. 15, 12489 Berlin, Germany
| | - Lari Lehtiö
- Faculty of Biochemistry and Molecular Medicine and Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Kresten Lindorff-Larsen
- Structural Biology and NMR Laboratory, Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - José Márquez
- European Molecular Biology Laboratory (EMBL) Grenoble, Grenoble, France
| | - André Matagne
- Laboratory of Enzymology and Protein Folding, Centre for Protein Engineering, InBioS Research Unit, University of Liège, Building B6C, Quartier Agora, Allée du 6 Août, 13, 4000 Liège (Sart-Tilman), Belgium
| | - Roberta Pierattelli
- Department of Chemistry "Ugo Schiff", University of Florence and Magnetic Resonance Center, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Antonio Rosato
- Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine-CIRMMP, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, Italy
| | - Frank Sobott
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Sridhar Sreeramulu
- Center for Biomolecular Magnetic Resonance (BMRZ), Institute for Organic Chemistry, Max-von-Laue-Str. 7, 60438 Frankfurt/M., Germany
| | - Jan Steyaert
- VIB-VUB Center for Structural Biology, VIB, Pleinlaan 2, Brussels, Belgium
| | - Joel L Sussman
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Lukas Trantirek
- Central European Institute of Technology (CEITEC), Masaryk University, Kamenice 753/5, 62500 Brno, Czech Republic
| | - Manfred S Weiss
- Macromolecular Crystallography, Helmholtz-Zentrum, Albert-Einstein-Str. 15, 12489 Berlin, Germany
| | - Matthias Wilmanns
- European Molecular Biology Laboratory (EMBL) Hamburg, Hamburg, Germany
| |
Collapse
|
9
|
Johnston TS, Hage C, Abedon AT, Panda S, Alejo JL, Eby Y, Segev DL, Tobian AAR, Cox AL, Werbel WA, Karaba AH. Rapid Wane and Recovery of XBB Sublineage Neutralization After Sequential Omicron-based Vaccination in Solid Organ Transplant Recipients. Clin Infect Dis 2024; 79:652-655. [PMID: 38953683 PMCID: PMC11426267 DOI: 10.1093/cid/ciae279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Indexed: 07/04/2024] Open
Abstract
Durability of variant neutralization in solid organ transplant recipients following Omicron-containing boosters is unknown. We report wane in XBB.1.5 neutralization by 3 months following a first bivalent booster, improved by a second booster; hybrid immunity improved peak, and duration of neutralization. Boosting at 3 to 6 months appears necessary to maintain neutralization.
Collapse
Affiliation(s)
- Trevor S Johnston
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Camille Hage
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aura T Abedon
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Snigdha Panda
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jennifer L Alejo
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yolanda Eby
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dorry L Segev
- Department of Surgery, NYU Grossman School of Medicine, New York, New York, USA
| | - Aaron A R Tobian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrea L Cox
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - William A Werbel
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew H Karaba
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
10
|
Meyers E, De Rop L, Engels F, Gioveni C, Coen A, De Burghgraeve T, Digregorio M, Van Ngoc P, De Clercq N, Buret L, Coenen S, Deschepper E, Padalko E, Callens S, Duysburgh E, De Sutter A, Scholtes B, Verbakel JY, Heytens S, Cools P. Follow-Up of SARS-CoV-2 Antibody Levels in Belgian Nursing Home Residents and Staff Two, Four and Six Months after Primary Course BNT162b2 Vaccination. Vaccines (Basel) 2024; 12:951. [PMID: 39204074 PMCID: PMC11359559 DOI: 10.3390/vaccines12080951] [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: 06/21/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/03/2024] Open
Abstract
When COVID-19 vaccines were implemented, nursing home residents (NHRs) and staff (NHS) in Belgium were prioritized for vaccination. To characterize the vaccine response over time in this population and to identify poorly responding groups, we assessed antibody concentrations two (T1), four (T2) and six months (T3) after primary course BNT162b2 vaccination in six groups of infection-naive/infection-primed NHRs/NHS, with/without comorbidity (NHRs only). Participant groups (N = 125 per group) were defined within a national serosurveillance study in nursing homes, based on questionnaire data. Dried blood spots were analyzed using ELISA for the quantification of SARS-CoV-2 S1RBD IgG antibodies. Among all groups, antibody concentrations significantly decreased between T1 and T2/T3, all with a ≥70% decrease at T3, except for infection-primed staff (-32%). Antibody concentrations among infection-naive NHRs were 11.96 times lower than those among infection-primed NHR, while the latter were comparable (x1.05) to infection-primed NHS. The largest proportion [13% (95% CI: 11-24%)] of vaccine non-responders was observed in the group of infection-naive NHRs with comorbidities. A longer interval between infection and vaccination (≥3 months) elicited higher antibody responses. Our data retrospectively show the necessity of timely COVID-19 booster vaccination. Infection-naive NHRs require special attention regarding immune monitoring in future epidemics or pandemics.
Collapse
Affiliation(s)
- Eline Meyers
- Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; (E.M.); (F.E.)
| | - Liselore De Rop
- EPI-Centre, Department of Public Health and Primary Care, KU Leuven, 3000 Leuven, Belgium
| | - Fien Engels
- Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; (E.M.); (F.E.)
| | - Claudia Gioveni
- Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; (E.M.); (F.E.)
| | - Anja Coen
- Department of Public Health and Primary Care, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium (S.H.)
| | - Tine De Burghgraeve
- EPI-Centre, Department of Public Health and Primary Care, KU Leuven, 3000 Leuven, Belgium
| | - Marina Digregorio
- Research Unit of Primary Care and Health, Department of General Medicine, Faculty of Medicine, University of Liège, 4000 Liège, Belgium
| | - Pauline Van Ngoc
- Research Unit of Primary Care and Health, Department of General Medicine, Faculty of Medicine, University of Liège, 4000 Liège, Belgium
| | - Nele De Clercq
- Department of Public Health and Primary Care, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium (S.H.)
| | - Laëtitia Buret
- Research Unit of Primary Care and Health, Department of General Medicine, Faculty of Medicine, University of Liège, 4000 Liège, Belgium
| | - Samuel Coenen
- Department of Family Medicine & Population Health, Faculty of Medicine and Health Sciences, University of Antwerp, 2000 Antwerp, Belgium
| | - Ellen Deschepper
- Biostatistics Unit, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Elizaveta Padalko
- Laboratory of Medical Microbiology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Steven Callens
- Department of Internal Medicine & Infectious Diseases, Ghent University Hospital, 9000 Ghent, Belgium
| | - Els Duysburgh
- Department of Epidemiology and Public Health, Sciensano, 1000 Brussels, Belgium
| | - An De Sutter
- Department of Public Health and Primary Care, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium (S.H.)
| | - Beatrice Scholtes
- Research Unit of Primary Care and Health, Department of General Medicine, Faculty of Medicine, University of Liège, 4000 Liège, Belgium
| | - Jan Y. Verbakel
- EPI-Centre, Department of Public Health and Primary Care, KU Leuven, 3000 Leuven, Belgium
| | - Stefan Heytens
- Department of Public Health and Primary Care, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium (S.H.)
| | - Piet Cools
- Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; (E.M.); (F.E.)
| |
Collapse
|
11
|
Hamm SR, Loft JA, Pérez-Alós L, Heftdal LD, Hansen CB, Møller DL, Pries-Heje MM, Hasselbalch RB, Fogh K, Hald A, Ostrowski SR, Frikke-Schmidt R, Sørensen E, Hilsted L, Bundgaard H, Garred P, Iversen K, Perch M, Sørensen SS, Rasmussen A, Sabin CA, Nielsen SD. The Impact of Time between Booster Doses on Humoral Immune Response in Solid Organ Transplant Recipients Vaccinated with BNT162b2 Vaccines. Viruses 2024; 16:860. [PMID: 38932153 PMCID: PMC11209529 DOI: 10.3390/v16060860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/19/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
As solid organ transplant (SOT) recipients remain at risk of severe outcomes after SARS-CoV-2 infections, vaccination continues to be an important preventive measure. In SOT recipients previously vaccinated with at least three doses of BNT162b2, we investigated humoral responses to BNT162b2 booster doses. Anti-SARS-CoV-2 receptor binding domain (RBD) immunoglobulin G (IgG) was measured using an in-house ELISA. Linear mixed models were fitted to investigate the change in the geometric mean concentration (GMC) of anti-SARS-CoV-2 RBD IgG after vaccination in participants with intervals of more or less than six months between the last two doses of vaccine. We included 107 SOT recipients vaccinated with a BNT162b2 vaccine. In participants with an interval of more than six months between the last two vaccine doses, we found a 1.34-fold change in GMC per month (95% CI 1.25-1.44), while we found a 1.09-fold change in GMC per month (95% CI 0.89-1.34) in participants with an interval of less than six months between the last two vaccine doses, resulting in a rate ratio of 0.82 (95% CI 0.66 to 1.01, p = 0.063). In conclusion, the administration of identical COVID-19 mRNA vaccine boosters within six months to SOT recipients may result in limited humoral immunogenicity of the last dose.
Collapse
Affiliation(s)
- Sebastian Rask Hamm
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Josefine Amalie Loft
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Laura Pérez-Alós
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen University Hospital, 2200 Copenhagen, Denmark
| | - Line Dam Heftdal
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Cecilie Bo Hansen
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen University Hospital, 2200 Copenhagen, Denmark
| | - Dina Leth Møller
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Mia Marie Pries-Heje
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Rasmus Bo Hasselbalch
- Department of Emergency Medicine, Herlev and Gentofte Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
- Department of Cardiology, Herlev and Gentofte Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Kamille Fogh
- Department of Emergency Medicine, Herlev and Gentofte Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
- Department of Cardiology, Herlev and Gentofte Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Annemette Hald
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Section 2034, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Erik Sørensen
- Department of Clinical Immunology, Section 2034, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Linda Hilsted
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Henning Bundgaard
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen University Hospital, 2200 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Kasper Iversen
- Department of Emergency Medicine, Herlev and Gentofte Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
- Department of Cardiology, Herlev and Gentofte Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Michael Perch
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Søren Schwartz Sørensen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Nephrology, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Allan Rasmussen
- Department of Surgical Gastroenterology, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Caroline A. Sabin
- Centre for Clinical Research, Epidemiology, Modelling and Evaluation, Institute for Global Health, UCL, Royal Free Campus, Rowland Hill St, London NW3 2PF, UK
| | - Susanne Dam Nielsen
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Surgical Gastroenterology, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| |
Collapse
|
12
|
Urschel R, Bronder S, Klemis V, Marx S, Hielscher F, Abu-Omar A, Guckelmus C, Schneitler S, Baum C, Becker SL, Gärtner BC, Sester U, Martinez L, Widera M, Schmidt T, Sester M. SARS-CoV-2-specific cellular and humoral immunity after bivalent BA.4/5 COVID-19-vaccination in previously infected and non-infected individuals. Nat Commun 2024; 15:3077. [PMID: 38594497 PMCID: PMC11004149 DOI: 10.1038/s41467-024-47429-8] [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: 06/12/2023] [Accepted: 04/02/2024] [Indexed: 04/11/2024] Open
Abstract
Knowledge is limited as to how prior SARS-CoV-2 infection influences cellular and humoral immunity after booster-vaccination with bivalent BA.4/5-adapted mRNA-vaccines, and whether vaccine-induced immunity may indicate subsequent infection. In this observational study, individuals with prior infection (n = 64) showed higher vaccine-induced anti-spike IgG-antibodies and neutralizing titers, but the relative increase was significantly higher in non-infected individuals (n = 63). In general, both groups showed higher neutralizing activity towards the parental strain than towards Omicron-subvariants BA.1, BA.2 and BA.5. In contrast, CD4 or CD8 T cell levels towards spike from the parental strain and the Omicron-subvariants, and cytokine expression profiles were similar irrespective of prior infection. Breakthrough infections occurred more frequently among previously non-infected individuals, who had significantly lower vaccine-induced spike-specific neutralizing activity and CD4 T cell levels. In summary, we show that immunogenicity after BA.4/5-bivalent vaccination differs between individuals with and without prior infection. Moreover, our results may help to improve prediction of breakthrough infections.
Collapse
Affiliation(s)
- Rebecca Urschel
- Department of Transplant and Infection Immunology, Saarland University, 66421, Homburg, Germany
| | - Saskia Bronder
- Department of Transplant and Infection Immunology, Saarland University, 66421, Homburg, Germany
| | - Verena Klemis
- Department of Transplant and Infection Immunology, Saarland University, 66421, Homburg, Germany
| | - Stefanie Marx
- Department of Transplant and Infection Immunology, Saarland University, 66421, Homburg, Germany
| | - Franziska Hielscher
- Department of Transplant and Infection Immunology, Saarland University, 66421, Homburg, Germany
| | - Amina Abu-Omar
- Department of Transplant and Infection Immunology, Saarland University, 66421, Homburg, Germany
| | - Candida Guckelmus
- Department of Transplant and Infection Immunology, Saarland University, 66421, Homburg, Germany
| | - Sophie Schneitler
- Institute of Medical Microbiology and Hygiene, Saarland University, 66421, Homburg, Germany
| | - Christina Baum
- Occupational Health Care Center, Saarland University, 66421, Homburg, Germany
| | - Sören L Becker
- Institute of Medical Microbiology and Hygiene, Saarland University, 66421, Homburg, Germany
| | - Barbara C Gärtner
- Institute of Medical Microbiology and Hygiene, Saarland University, 66421, Homburg, Germany
| | - Urban Sester
- Department of Nephrology, SHG-Klinikum Völklingen, 66333, Völklingen, Germany
| | - Leonardo Martinez
- Boston University, School of Public Health, Department of Epidemiology, Boston, MA, USA
| | - Marek Widera
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Tina Schmidt
- Department of Transplant and Infection Immunology, Saarland University, 66421, Homburg, Germany
| | - Martina Sester
- Department of Transplant and Infection Immunology, Saarland University, 66421, Homburg, Germany.
- Center for Gender-specific Biology and Medicine (CGBM), Saarland University, 66421, Homburg, Germany.
| |
Collapse
|
13
|
Goguet E, Olsen CH, Meyer WA, Ansari S, Powers JH, Conner TL, Coggins SA, Wang W, Wang R, Illinik L, Sanchez Edwards M, Jackson-Thompson BM, Hollis-Perry M, Wang G, Alcorta Y, Wong MA, Saunders D, Mohammed R, Balogun B, Kobi P, Kosh L, Bishop-Lilly K, Cer RZ, Arnold CE, Voegtly LJ, Fitzpatrick M, Luquette AE, Malagon F, Ortega O, Parmelee E, Davies J, Lindrose AR, Haines-Hull H, Moser MS, Samuels EC, Rekedal MS, Graydon EK, Malloy AMW, Tribble D, Burgess TH, Campbell W, Robinson S, Broder CC, O’Connell RJ, Weiss CD, Pollett S, Laing E, Mitre E. Immune and behavioral correlates of protection against symptomatic post-vaccination SARS-CoV-2 infection. Front Immunol 2024; 15:1287504. [PMID: 38566991 PMCID: PMC10985347 DOI: 10.3389/fimmu.2024.1287504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 02/27/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction We sought to determine pre-infection correlates of protection against SARS-CoV-2 post-vaccine inzfections (PVI) acquired during the first Omicron wave in the United States. Methods Serum and saliva samples from 176 vaccinated adults were collected from October to December of 2021, immediately before the Omicron wave, and assessed for SARS-CoV-2 Spike-specific IgG and IgA binding antibodies (bAb). Sera were also assessed for bAb using commercial assays, and for neutralization activity against several SARS-CoV-2 variants. PVI duration and severity, as well as risk and precautionary behaviors, were assessed by questionnaires. Results Serum anti-Spike IgG levels assessed by research assay, neutralization titers against Omicron subvariants, and low home risk scores correlated with protection against PVIs after multivariable regression analysis. Commercial assays did not perform as well as research assay, likely due to their lower dynamic range. Discussion In the 32 participants that developed PVI, anti-Spike IgG bAbs correlated with lower disease severity and shorter duration of illness.
Collapse
Affiliation(s)
- Emilie Goguet
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Cara H. Olsen
- Department of Preventive Medicine & Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | | | - Sara Ansari
- Quest Diagnostics, Secaucus, NJ, United States
| | - John H. Powers
- Clinical Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Tonia L. Conner
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Si’Ana A. Coggins
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Wei Wang
- Division of Viral Products, Office of Vaccine Research and Review, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Richard Wang
- Division of Viral Products, Office of Vaccine Research and Review, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Luca Illinik
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Margaret Sanchez Edwards
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Belinda M. Jackson-Thompson
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Monique Hollis-Perry
- Clinical Trials Center, Infectious Diseases Directorate, Naval Medical Research Center, Silver Spring, MD, United States
| | - Gregory Wang
- Clinical Trials Center, Infectious Diseases Directorate, Naval Medical Research Center, Silver Spring, MD, United States
- General Dynamics Information Technology, Falls Church, VA, United States
| | - Yolanda Alcorta
- Clinical Trials Center, Infectious Diseases Directorate, Naval Medical Research Center, Silver Spring, MD, United States
- General Dynamics Information Technology, Falls Church, VA, United States
| | - Mimi A. Wong
- Clinical Trials Center, Infectious Diseases Directorate, Naval Medical Research Center, Silver Spring, MD, United States
- General Dynamics Information Technology, Falls Church, VA, United States
| | - David Saunders
- Translational Medicine Unit, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Roshila Mohammed
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Bolatito Balogun
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Priscilla Kobi
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Lakeesha Kosh
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Kimberly Bishop-Lilly
- Biological Defense Research Directorate, Naval Medical Research Command, Fort Detrick, MD, United States
| | - Regina Z. Cer
- Biological Defense Research Directorate, Naval Medical Research Command, Fort Detrick, MD, United States
| | - Catherine E. Arnold
- Biological Defense Research Directorate, Naval Medical Research Command, Fort Detrick, MD, United States
- Defense Threat Reduction Agency, Fort Belvoir, VA, United States
| | - Logan J. Voegtly
- Biological Defense Research Directorate, Naval Medical Research Command, Fort Detrick, MD, United States
- Leidos, Reston, VA, United States
| | - Maren Fitzpatrick
- Biological Defense Research Directorate, Naval Medical Research Command, Fort Detrick, MD, United States
- Leidos, Reston, VA, United States
| | - Andrea E. Luquette
- Biological Defense Research Directorate, Naval Medical Research Command, Fort Detrick, MD, United States
- Leidos, Reston, VA, United States
| | - Francisco Malagon
- Biological Defense Research Directorate, Naval Medical Research Command, Fort Detrick, MD, United States
- Leidos, Reston, VA, United States
| | - Orlando Ortega
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Edward Parmelee
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Julian Davies
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Alyssa R. Lindrose
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Hannah Haines-Hull
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Matthew S. Moser
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Emily C. Samuels
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Marana S. Rekedal
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Elizabeth K. Graydon
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Allison M. W. Malloy
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - David R. Tribble
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Timothy H. Burgess
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Wesley Campbell
- Division of Infectious Diseases, Walter Reed National Military Medical Center, Bethesda, MD, United States
| | - Sara Robinson
- Division of Infectious Diseases, Walter Reed National Military Medical Center, Bethesda, MD, United States
| | - Christopher C. Broder
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Robert J. O’Connell
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Carol D. Weiss
- Division of Viral Products, Office of Vaccine Research and Review, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Simon Pollett
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Eric D. Laing
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Edward Mitre
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
14
|
Leye E, Delory T, El Karoui K, Espagnacq M, Khlat M, Le Coeur S, Lapidus N, Hejblum G. Direct and indirect impact of the COVID-19 pandemic on the survival of kidney transplant recipients: A national observational study in France. Am J Transplant 2024; 24:479-490. [PMID: 37898317 DOI: 10.1016/j.ajt.2023.10.017] [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: 05/25/2023] [Revised: 09/28/2023] [Accepted: 10/20/2023] [Indexed: 10/30/2023]
Abstract
During the pandemic period, health care systems were substantially reorganized for managing COVID-19 cases. Corresponding consequences on persons with chronic diseases remain insufficiently documented. This observational cohort study investigated the direct and indirect impact of the pandemic period on the survival of kidney transplant recipients (KTR). Using the French National Health Data System, incident persons with end-stage kidney disease between 2015 and 2020, and who received a kidney transplant during this period were included and followed up from their transplantation date to December 31, 2021. The survival of KTR during the prepandemic and pandemic periods was investigated using Cox models with time-dependent covariates. There were 10 637 KTR included in the study, with 324 and 430 deaths observed during the prepandemic and pandemic periods, respectively. The adjusted risk of death during the pandemic period was similar to that observed during the prepandemic period (hazard ratio [HR] [95% confidence interval]: 0.92 [0.77-1.11]), COVID-19-related hospitalization was associated with an increased risk of death (HR: 10.62 [8.46-13.33]), and a third vaccine dose was associated with a lower risk of death (HR: 0.42 [0.30-0.57]). The pandemic period was not associated with an indirect higher risk of death in KTR with no COVID-19-related hospitalization.
Collapse
Affiliation(s)
- Elhadji Leye
- Sorbonne Université, INSERM, Institut Pierre Louis d'Épidémiologie et de Santé Publique, Paris, France.
| | - Tristan Delory
- Sorbonne Université, INSERM, Institut Pierre Louis d'Épidémiologie et de Santé Publique, Paris, France; Centre Hospitalier Annecy-Genevois, Epagny Metz-Tessy, France; French Institute for Demographic Studies (INED), Mortality, Health and Epidemiology Unit, Aubervilliers, France
| | - Khalil El Karoui
- Sorbonne Université, INSERM U1155, AP-HP, Hôpital Tenon, Service de Néphrologie, Paris, France
| | - Maude Espagnacq
- Institute for Research and Information in Health Economics (IRDES), Paris, France
| | - Myriam Khlat
- French Institute for Demographic Studies (INED), Mortality, Health and Epidemiology Unit, Aubervilliers, France
| | - Sophie Le Coeur
- French Institute for Demographic Studies (INED), Mortality, Health and Epidemiology Unit, Aubervilliers, France
| | - Nathanaël Lapidus
- Sorbonne Université, INSERM, Institut Pierre Louis d'Épidémiologie et de Santé Publique, AP-HP, Hôpital Saint-Antoine, Unité de Santé Publique, Paris, France
| | - Gilles Hejblum
- Sorbonne Université, INSERM, Institut Pierre Louis d'Épidémiologie et de Santé Publique, Paris, France
| |
Collapse
|
15
|
Han A, Min S, Jo EA, Lee H, Kim YC, Han SS, Kang HG, Ahn YH, Oh I, Song EY, Ha J. Association Between Low Anti-spike Antibody Levels After the Third Dose of SARS-CoV-2 Vaccination and Hospitalization due to Symptomatic Breakthrough Infection in Kidney Transplant Recipients. Ann Lab Med 2024; 44:64-73. [PMID: 37665287 PMCID: PMC10485855 DOI: 10.3343/alm.2024.44.1.64] [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: 03/27/2023] [Revised: 05/11/2023] [Accepted: 07/26/2023] [Indexed: 09/05/2023] Open
Abstract
Background Whether anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibody levels post-third coronavirus disease (COVID-19) vaccination correlate with worse outcomes due to breakthrough infection is unclear. We evaluated the association between anti-SARS-CoV-2 antibody levels and symptomatic breakthrough infection or hospitalization during the Omicron surge in kidney transplant recipients. Methods In total, 287 kidney transplant recipients expected to receive a third vaccination were enrolled between November 2021 and February 2022. The Abbott SARS-CoV-2 IgG II Quant test (Abbott, Chicago, IL, USA) was performed within three weeks before and four weeks after the third vaccination. The incidence of symptomatic breakthrough infection and hospitalization from two weeks to four months post-third vaccination was recorded. Results After the third vaccination, the seropositive rate and median antibody titer of the 287 patients increased from 57.1% to 82.2% and from 71.7 (interquartile range [IQR] 7.2-402.8) to 1,612.1 (IQR 153.9-5,489.1) AU/mL, respectively. Sixty-four (22.3%) patients had symptomatic breakthrough infections, of whom 12 required hospitalization. Lower anti-receptor-binding domain (RBD) IgG levels (<400 AU/mL) post-third vaccination were a risk factor for symptomatic breakthrough infection (hazard ratio [HR]=3.46, P<0.001). Anti-RBD IgG levels <200 AU/mL were a critical risk factor for hospitalization (HR=36.4, P=0.007). Conclusions Low anti-spike IgG levels after third vaccination in kidney transplant recipients were associated with symptomatic breakthrough infection and, particularly, with hospitalization during the Omicron surge. These data can be used to identify patients requiring additional protective measures, such as passive immunization using monoclonal antibodies.
Collapse
Affiliation(s)
- Ahram Han
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Sangil Min
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Eun-Ah Jo
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Hajeong Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Yong Chul Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Seung Seok Han
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Hee Gyung Kang
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | - Yo Han Ahn
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | - Inseong Oh
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Eun Young Song
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jongwon Ha
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
- Transplantation Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
16
|
Kang H, Oh EJ. New Insights Into SARS-CoV-2-specific Antibody Levels in Kidney Transplantation Recipients After Three Vaccination Doses. Ann Lab Med 2024; 44:3-5. [PMID: 37665280 PMCID: PMC10485850 DOI: 10.3343/alm.2024.44.1.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023] Open
Affiliation(s)
- Hyunhye Kang
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Eun-Jee Oh
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
17
|
Yap DYH, Fong CHY, Zhang X, Ip JD, Chan WM, Chu AWH, Chen LL, Zhao Y, Chan BPC, Luk KS, Cheng VCC, Chan TM, To KKW. Humoral and cellular immunity against different SARS-CoV-2 variants in patients with chronic kidney disease. Sci Rep 2023; 13:19932. [PMID: 37968273 PMCID: PMC10652016 DOI: 10.1038/s41598-023-47130-8] [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: 08/11/2023] [Accepted: 11/09/2023] [Indexed: 11/17/2023] Open
Abstract
Chronic kidney disease (CKD) patients are at higher risk of severe COVID-19. Humoral and cellular immunity from prior infection or vaccination are important for protection, but the neutralizing antibody (nAb) response against SARS-CoV-2 variants is impaired. We investigated the variant-specific nAb and T cell immunity among CKD patients. Adult CKD patients were recruited between August and October 2022. nAb against the SARS-CoV-2 (ancestral strains and four Omicron sublineages) and T cell response were measured using the live virus neutralization assay and interferon-gamma release assay (IGRA). The correlation between nAb/T-cell response and subsequent infection after recruitment were also determined. Among the 88 recruited patients, 95.5% had prior infection or had completed the primary vaccine series. However, only 77.3% had detectable nAb against at least one SARS-CoV-2 strains, 59.1% tested positive in IGRA, and 52.3% had detectable nAb and tested positive in the IGRA. The nAb geometic mean titers (GMTs) against XBB.1, BA.5 and BA.2.3.20 were significantly lower than those against BA.2 and ancestral strain. Prior SARS-CoV-2 infection was associated with elevated nAb and T cell response. More kidney transplant recipients (KTRs) showed absent nAb and T cell response (36.8% vs. 10.1%), despite a higher prevalence of vaccine booster in this population (94.7% vs. 50.7%). Lower levels of nAb titer and T cell response were significantly associated with subsequent infection. A considerable proportion of CKD patients, especially KTRs, showed absence of humoral and cellular protective immunity against SARS-CoV-2. Strategies to improve immunogenicity in this population are urgently needed.
Collapse
Affiliation(s)
- Desmond Yat-Hin Yap
- Division of Nephrology, Department of Medicine, Queen Mary Hospital, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Carol Ho-Yan Fong
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Shatin, Hong Kong Special Administrative Region, People's Republic of China
| | - Xiaojuan Zhang
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Jonathan Daniel Ip
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Wan-Mui Chan
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Allen Wing-Ho Chu
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Shatin, Hong Kong Special Administrative Region, People's Republic of China
| | - Lin-Lei Chen
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Yan Zhao
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Brian Pui-Chun Chan
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Kristine Shik Luk
- Department of Pathology, Princess Margaret Hospital, Kwai Chung, Hong Kong Special Administrative Region, People's Republic of China
| | - Vincent Chi-Chung Cheng
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Tak-Mao Chan
- Division of Nephrology, Department of Medicine, Queen Mary Hospital, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Kelvin Kai-Wang To
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Shatin, Hong Kong Special Administrative Region, People's Republic of China.
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, People's Republic of China.
| |
Collapse
|
18
|
Udomkarnjananun S, Gatechompol S, Leelahavanichkul A, Kerr SJ. Cellular immune response of SARS-CoV-2 vaccination in kidney transplant recipients: a systematic review and meta-analysis. Front Immunol 2023; 14:1220148. [PMID: 37575225 PMCID: PMC10415203 DOI: 10.3389/fimmu.2023.1220148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/05/2023] [Indexed: 08/15/2023] Open
Abstract
Background Evidence has demonstrated inferior humoral immune responses after SARS-CoV-2 vaccination in kidney transplant recipients compared to the general population. However, data on cellular immune responses in this population have not been established. Methods We searched the MEDLINE, Scopus, and Cochrane databases and included studies reporting cellular immune response rates in kidney transplant recipients after receiving SARS-CoV-2 vaccines. Studies that reported factors associated with cellular immune responders or non-responders were also included (PROSPERO: CRD42022375544). Results From a total of 1,494 articles searched, 53 articles were included in the meta-analysis. In all, 21 studies assessed cellular immune response by interferon-γ enzyme-linked immunosorbent spot (IFN-γ ELISPOT), 22 studies used interferon-γ release assay (IGRA), and 10 studies used flow cytometric analysis. The pooled response rate after two doses (standard regimen) and three doses of vaccination was 47.5% (95%CI 38.4-56.7%) and 69.1% (95%CI 56.3-80.6%) from IFN-γ ELISPOT, 25.8% (95%CI 19.7-32.4%) and 14.7% (95%CI 8.5-22.2%) from IGRA, and 73.7% (95%CI 55.2-88.8%) and 86.5% (95%CI 75.3-94.9%) from flow cytometry, respectively. Recipients with seroconversion were associated with a higher chance of having cellular immune response (OR 2.58; 95%CI 1.89-3.54). Cellular immune response in kidney transplant recipients was lower than in dialysis patients (OR 0.24; 95%CI 0.16-0.34) and the general population (OR 0.10; 95%CI 0.07-0.14). Age and immunosuppressants containing tacrolimus or corticosteroid were associated with inferior cellular immune response. Conclusion Cellular immune response after SARS-CoV-2 vaccination in kidney transplant recipients was lower than in dialysis patients and the general population. Age, tacrolimus, and corticosteroid were associated with poor response. Cellular immune response should also be prioritized in vaccination studies. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42022375544.
Collapse
Affiliation(s)
- Suwasin Udomkarnjananun
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
- Excellence Center for Organ Transplantation (ECOT), King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Renal Immunology and Transplantation Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Asada Leelahavanichkul
- Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Chulalongkorn University, Bangkok, Thailand
- Immunology Unit, Department of Microbiology, Chulalongkorn University, Bangkok, Thailand
| | - Stephen J. Kerr
- HIV-NAT, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
- Biostatistics Excellence Centre, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- The Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
19
|
Federico L, Tvedt THA, Gainullin M, Osen JR, Chaban V, Lund KP, Tietze L, Tran TT, Lund-Johansen F, Kared H, Lind A, Vaage JT, Stratford R, Tennøe S, Malone B, Clancy T, Myhre AEL, Gedde-Dahl T, Munthe LA. Robust spike-specific CD4 + and CD8 + T cell responses in SARS-CoV-2 vaccinated hematopoietic cell transplantation recipients: a prospective, cohort study. Front Immunol 2023; 14:1210899. [PMID: 37503339 PMCID: PMC10369799 DOI: 10.3389/fimmu.2023.1210899] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/13/2023] [Indexed: 07/29/2023] Open
Abstract
Poor overall survival of hematopoietic stem cell transplantation (HSCT) recipients who developed COVID-19 underlies the importance of SARS-CoV-2 vaccination. Previous studies of vaccine efficacy have reported weak humoral responses but conflicting results on T cell immunity. Here, we have examined the relationship between humoral and T cell response in 48 HSCT recipients who received two doses of Moderna's mRNA-1273 or Pfizer/BioNTech's BNT162b2 vaccines. Nearly all HSCT patients had robust T cell immunity regardless of protective humoral responses, with 18/48 (37%, IQR 8.679-5601 BAU/mL) displaying protective IgG anti-receptor binding domain (RBD) levels (>2000 BAU/mL). Flow cytometry analysis of activation induced markers (AIMs) revealed that 90% and 74% of HSCT patients showed reactivity towards immunodominant spike peptides in CD8+ and CD4+ T cells, respectively. The response rate increased to 90% for CD4+ T cells as well when we challenged the cells with a complete set of overlapping peptides spanning the entire spike protein. T cell response was detectable as early as 3 months after transplant, but only CD4+ T cell reactivity correlated with IgG anti-RBD level and time after transplantation. Boosting increased seroconversion rate, while only one patient developed COVID-19 requiring hospitalization. Our data suggest that HSCT recipients with poor serological responses were protected from severe COVID-19 by vaccine-induced T cell responses.
Collapse
Affiliation(s)
- Lorenzo Federico
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Murat Gainullin
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Julie Røkke Osen
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Viktoriia Chaban
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Katrine Persgård Lund
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Lisa Tietze
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- ImmunoLingo Convergence Center, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Trung The Tran
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- ImmunoLingo Convergence Center, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- ImmunoLingo Convergence Center, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Hassen Kared
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Andreas Lind
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - John Torgils Vaage
- ImmunoLingo Convergence Center, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | | | | | | | - Anders Eivind Leren Myhre
- Department of Haematology, Oslo University Hospital, Oslo, Norway
- ImmunoLingo Convergence Center, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Ludvig André Munthe
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
20
|
Del Mastro A, Picascia S, D'Apice L, Trovato M, Barba P, Di Biase I, Di Biase S, Laccetti M, Belli A, Amato G, Di Muro P, Credendino O, Picardi A, De Berardinis P, Del Pozzo G, Gianfrani C. Booster Dose of SARS-CoV-2 mRNA Vaccine in Kidney Transplanted Patients Induces Wuhan-Hu-1 Specific Neutralizing Antibodies and T Cell Activation but Lower Response against Omicron Variant. Viruses 2023; 15:v15051132. [PMID: 37243218 DOI: 10.3390/v15051132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/29/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Kidney transplanted recipients (KTR) are at high risk of severe SARS-CoV-2 infection due to immunosuppressive therapy. Although several studies reported antibody production in KTR after vaccination, data related to immunity to the Omicron (B.1.1.529) variant are sparse. Herein, we analyzed anti-SARS-CoV-2 immune response in seven KTR and eight healthy controls after the second and third dose of the mRNA vaccine (BNT162b2). A significant increase in neutralizing antibody (nAb) titers were detected against pseudoviruses expressing the Wuhan-Hu-1 spike (S) protein after the third dose in both groups, although nAbs in KTR were lower than controls. nAbs against pseudoviruses expressing the Omicron S protein were low in both groups, with no increase after the 3rd dose in KTR. Reactivity of CD4+ T cells after boosting was observed when cells were challenged with Wuhan-Hu-1 S peptides, while Omicron S peptides were less effective in both groups. IFN-γ production was detected in KTR in response to ancestral S peptides, confirming antigen-specific T cell activation. Our study demonstrates that the 3rd mRNA dose induces T cell response against Wuhan-Hu-1 spike peptides in KTR, and an increment in the humoral immunity. Instead, humoral and cellular immunity to Omicron variant immunogenic peptides were low in both KTR and healthy vaccinated subjects.
Collapse
Affiliation(s)
- Andrea Del Mastro
- AORN A. Cardarelli-Internal Medicine Division 1-Immunology Unit, 80131 Naples, Italy
| | - Stefania Picascia
- Institute of Biochemistry and Cell Biology, Italian National Council of Research, 80131 Naples, Italy
| | - Luciana D'Apice
- Institute of Biochemistry and Cell Biology, Italian National Council of Research, 80131 Naples, Italy
| | - Maria Trovato
- Institute of Biochemistry and Cell Biology, Italian National Council of Research, 80131 Naples, Italy
| | - Pasquale Barba
- Institute of Biochemistry and Cell Biology, Italian National Council of Research, 80131 Naples, Italy
| | | | | | - Marco Laccetti
- AORN A. Cardarelli-Internal Medicine Division 1-Immunology Unit, 80131 Naples, Italy
| | - Antonello Belli
- AORN A. Cardarelli-Clinical Pathology Division, 80131 Naples, Italy
| | - Gerardino Amato
- AORN A. Cardarelli-Clinical Pathology Division, 80131 Naples, Italy
| | - Potito Di Muro
- AORN A. Cardarelli-Nephrology and Dialysis Unit, 80131 Naples, Italy
| | - Olga Credendino
- AORN A. Cardarelli-Nephrology and Dialysis Unit, 80131 Naples, Italy
| | - Alessandra Picardi
- AORN A. Cardarelli-Molecular Biology Laboratory-Hematology and HSC Transplantation Unit, 80131 Naples, Italy
| | | | - Giovanna Del Pozzo
- Institute of Genetics and Biophysics, Italian National Council of Research, 80131 Naples, Italy
| | - Carmen Gianfrani
- Institute of Biochemistry and Cell Biology, Italian National Council of Research, 80131 Naples, Italy
| |
Collapse
|
21
|
Cromer D, Steain M, Reynaldi A, Schlub TE, Khan SR, Sasson SC, Kent SJ, Khoury DS, Davenport MP. Predicting vaccine effectiveness against severe COVID-19 over time and against variants: a meta-analysis. Nat Commun 2023; 14:1633. [PMID: 36964146 PMCID: PMC10036966 DOI: 10.1038/s41467-023-37176-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 03/06/2023] [Indexed: 03/26/2023] Open
Abstract
Vaccine protection from symptomatic SARS-CoV-2 infection has been shown to be strongly correlated with neutralising antibody titres; however, this has not yet been demonstrated for severe COVID-19. To explore whether this relationship also holds for severe COVID-19, we performed a systematic search for studies reporting on protection against different SARS-CoV-2 clinical endpoints and extracted data from 15 studies. Since matched neutralising antibody titres were not available, we used the vaccine regimen, time since vaccination and variant of concern to predict corresponding neutralising antibody titres. We then compared the observed vaccine effectiveness reported in these studies to the protection predicted by a previously published model of the relationship between neutralising antibody titre and vaccine effectiveness against severe COVID-19. We find that predicted neutralising antibody titres are strongly correlated with observed vaccine effectiveness against symptomatic (Spearman [Formula: see text] = 0.95, p < 0.001) and severe (Spearman [Formula: see text] = 0.72, p < 0.001 for both) COVID-19 and that the loss of neutralising antibodies over time and to new variants are strongly predictive of observed vaccine protection against severe COVID-19.
Collapse
Affiliation(s)
- Deborah Cromer
- Kirby Institute, University of New South Wales, Sydney, Australia.
| | - Megan Steain
- Sydney Institute of Infectious Diseases and Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Timothy E Schlub
- Kirby Institute, University of New South Wales, Sydney, Australia
- Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Shanchita R Khan
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Sarah C Sasson
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
| | - David S Khoury
- Kirby Institute, University of New South Wales, Sydney, Australia
| | | |
Collapse
|