1
|
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 2024; 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] [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
|
2
|
Kinsella PM, Moso MA, Morrissey CO, Dendle C, Guy S, Bond K, Sasadeusz J, Slavin MA. Antiviral therapies for the management of persistent coronavirus disease 2019 in immunocompromised hosts: A narrative review. Transpl Infect Dis 2024; 26:e14301. [PMID: 38809102 DOI: 10.1111/tid.14301] [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: 12/10/2023] [Revised: 04/11/2024] [Accepted: 05/08/2024] [Indexed: 05/30/2024]
Abstract
Antiviral agents with activity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have played a critical role in disease management; however, little is known regarding the efficacy of these medications in the treatment of SARS-CoV-2 infection in immunocompromised patients, particularly in the management of persistent SARS-CoV-2 positivity. This narrative review discusses the management of persistent coronavirus disease 2019 in immunocompromised hosts, with a focus on antiviral therapies. We identified 84 cases from the literature describing a variety of approaches, including prolonged antiviral therapy (n = 11), combination antivirals (n = 13), and mixed therapy with antiviral and antibody treatments (n = 60). A high proportion had an underlying haematologic malignancy (n = 67, 80%), and were in receipt of anti-CD20 agents (n = 51, 60%). Success was reported in 70 cases (83%) which varied according to the therapy type. Combination therapies with antivirals may be an effective approach for individuals with persistent SARS-CoV-2 positivity, particularly those that incorporate treatments aimed at increasing neutralizing antibody levels. Any novel approaches taken to this difficult management dilemma should be mindful of the emergence of antiviral resistance.
Collapse
Affiliation(s)
- Paul M Kinsella
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Infectious Diseases, University of Melbourne at the Doherty Institute of Infection and Immunity, Melbourne, Australia
| | - Michael A Moso
- Department of Infectious Diseases, University of Melbourne at the Doherty Institute of Infection and Immunity, Melbourne, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Doherty Institute of Infection and Immunity, Melbourne, Australia
| | | | - Claire Dendle
- Monash Infectious Diseases, Monash Health, Melbourne, Australia
- School of Clinical Sciences, Monash University, Melbourne, Australia
| | - Stephen Guy
- Department of Infectious Diseases, Eastern Health, Melbourne, Australia
- Eastern Health Clinical School, Monash University, Melbourne, Australia
| | - Katherine Bond
- Department of Microbiology, Royal Melbourne Hospital, Melbourne, Australia
- Victorian Infectious Diseases Reference Laboratory (VIDRL) at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Microbiology and Immunology, University of Melbourne at the Doherty Institute of Infection and Immunity, Melbourne, Australia
| | - Joseph Sasadeusz
- Department of Infectious Diseases, University of Melbourne at the Doherty Institute of Infection and Immunity, Melbourne, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Doherty Institute of Infection and Immunity, Melbourne, Australia
| | - Monica A Slavin
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Doherty Institute of Infection and Immunity, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| |
Collapse
|
3
|
Dandu H, Goel A, Kumar M, Malhotra HS, Katiyar H, Agarwal M, Kumar N, Pandey P, Rani S, Yadav G. Humoral and cellular immune response in patients of liver cirrhosis and immunocompetent recipient of ChAdOx1nCoV-19 Vaccine (Covishield). Clin Exp Med 2024; 24:24. [PMID: 38280060 PMCID: PMC10821839 DOI: 10.1007/s10238-023-01258-z] [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: 09/11/2023] [Accepted: 12/15/2023] [Indexed: 01/29/2024]
Abstract
Despite the effectiveness of COVID-19 vaccination in reducing the severity of the disease, the demand for booster is increasing in vulnerable populations like elderly and immunocompromised individuals especially with each new wave of COVID-19 in different countries. There is limited data on the sustained immunity against COVID-19 in patients with liver cirrhosis. The study was aimed to compare the T cell and humoral immune response after 1 year of ChAdOx1nCoV-19 Vaccine in patients with liver cirrhosis and healthy health care workers (HCW). This was a prospective observational study including 36 HCW, 19 liver cirrhosis patients and 10 unvaccinated individuals. Anti-SARS-CoV-2S antibody, neutralizing antibody and memory T cell subsets were evaluated by ELISA and flow cytometry, respectively, in all three groups after 1 year of initial vaccination. Compared to HCW and unvaccinated individuals, liver cirrhosis patients had significantly depleted T cells, although CD4:CD8 + T cell ratio was normal. Both cirrhotic patients and HCW developed memory T cell subset [effector memory RA (P = 0.141, P < 0.001), effector memory (P < 0.001, P < 0.001), central memory (P < 0.001, P < 0.01), stem cell memory (P = 0.009, P = 0.08) and naïve (P < 0.001, P = 0.02)] compared to unvaccinated unexposed individuals of CD4 + T and CD8 + T, respectively. However, among HCW and cirrhotic group no difference was noted on central memory and stem cell memory cells on T cells. Patients with liver cirrhosis developed comparable memory T cells after vaccination which can evoke sustainable immune response on reinfection. Therefore, additional vaccine doses may not be necessary for cirrhosis patients.
Collapse
Affiliation(s)
- Himanshu Dandu
- Department of Internal Medicine, King George's Medical University, Lucknow, 226003, India
| | - Amit Goel
- Department of Hepatology, Sanjay Gandhi Post-Graduate Institute of Medical Sciences, Lucknow, 226014, India
| | - Manish Kumar
- Department of Pathology, King George's Medical University, Lucknow, 226003, India
| | | | - Harshita Katiyar
- Department of Hepatology, Sanjay Gandhi Post-Graduate Institute of Medical Sciences, Lucknow, 226014, India
| | - Monica Agarwal
- Department of Community Medicine, King George's Medical University, Lucknow, 226003, India
| | - Neeraj Kumar
- Department of Neurology, King George's Medical University, Lucknow, 226003, India
| | - Pragya Pandey
- Department of Conservative Dentistry and Endodontics, King George's Medical University, Lucknow, 226003, India
| | - Shivani Rani
- Department of Internal Medicine, King George's Medical University, Lucknow, 226003, India
| | - Geeta Yadav
- Department of Pathology, King George's Medical University, Lucknow, 226003, India.
| |
Collapse
|
4
|
Alexander KL, Ford ML. The Entangled World of Memory T Cells and Implications in Transplantation. Transplantation 2024; 108:137-147. [PMID: 37271872 PMCID: PMC10696133 DOI: 10.1097/tp.0000000000004647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Memory T cells that are specific for alloantigen can arise from a variety of stimuli, ranging from direct allogeneic sensitization from prior transplantation, blood transfusion, or pregnancy to the elicitation of pathogen-specific T cells that are cross-reactive with alloantigen. Regardless of the mechanism by which they arise, alloreactive memory T cells possess key metabolic, phenotypic, and functional properties that render them distinct from naive T cells. These properties affect the immune response to transplantation in 2 important ways: first, they can alter the speed, location, and effector mechanisms with which alloreactive T cells mediate allograft rejection, and second, they can alter T-cell susceptibility to immunosuppression. In this review, we discuss recent developments in understanding these properties of memory T cells and their implications for transplantation.
Collapse
Affiliation(s)
| | - Mandy L. Ford
- Emory Transplant Center, Emory University, Atlanta, GA
| |
Collapse
|
5
|
Rendina M, Barone M, Lillo C, Trapani S, Masiero L, Trerotoli P, Puoti F, Lupo LG, Tandoi F, Agnes S, Grieco A, Andorno E, Marenco S, Giannini EG, Baccarani U, Toniutto P, Carraro A, Colecchia A, Cescon M, Morelli MC, Cillo U, Burra P, Angeli P, Colledan M, Fagiuoli S, De Carlis L, Belli L, De Simone P, Carrai P, Di Benedetto F, De Maria N, Ettorre GM, Giannelli V, Gruttadauria S, Volpes R, Corsale S, Mazzaferro V, Bhoori S, Romagnoli R, Martini S, Rossi G, Caccamo L, Donato MF, Rossi M, Ginanni Corradini S, Spada M, Maggiore G, Tisone G, Lenci I, Vennarecci G, Tortora R, Vivarelli M, Svegliati Baroni G, Zamboni F, Mameli L, Tafuri S, Simone S, Gesualdo L, Cardillo M, Di Leo A. The Italian data on SARS-CoV-2 infection in transplanted patients support an organ specific immune response in liver recipients. Front Immunol 2023; 14:1203854. [PMID: 37469512 PMCID: PMC10352984 DOI: 10.3389/fimmu.2023.1203854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/18/2023] [Indexed: 07/21/2023] Open
Abstract
Introduction The study of immune response to SARSCoV-2 infection in different solid organ transplant settings represents an opportunity for clarifying the interplay between SARS-CoV-2 and the immune system. In our nationwide registry study from Italy, we specifically evaluated, during the first wave pandemic, i.e., in non-vaccinated patients, COVID-19 prevalence of infection, mortality, and lethality in liver transplant recipients (LTRs), using non-liver solid transplant recipients (NL-SOTRs) and the Italian general population (GP) as comparators. Methods Case collection started from February 21 to June 22, 2020, using the data from the National Institute of Health and National Transplant Center, whereas the data analysis was performed on September 30, 2020.To compare the sex- and age-adjusted distribution of infection, mortality, and lethality in LTRs, NL-SOTRs, and Italian GP we applied an indirect standardization method to determine the standardized rate. Results Among the 43,983 Italian SOTRs with a functioning graft, LTRs accounted for 14,168 patients, of whom 89 were SARS-CoV-2 infected. In the 29,815 NL-SOTRs, 361 cases of SARS-CoV-2 infection were observed. The geographical distribution of the disease was highly variable across the different Italian regions. The standardized rate of infection, mortality, and lethality rates in LTRs resulted lower compared to NL-SOTRs [1.02 (95%CI 0.81-1.23) vs. 2.01 (95%CI 1.8-2.2); 1.0 (95%CI 0.5-1.5) vs. 4.5 (95%CI 3.6-5.3); 1.6 (95%CI 0.7-2.4) vs. 2.8 (95%CI 2.2-3.3), respectively] and comparable to the Italian GP. Discussion According to the most recent studies on SOTRs and SARS-CoV-2 infection, our data strongly suggest that, in contrast to what was observed in NL-SOTRs receiving a similar immunosuppressive therapy, LTRs have the same risk of SARS-CoV-2 infection, mortality, and lethality observed in the general population. These results suggest an immune response to SARS-CoV-2 infection in LTRS that is different from NL-SOTRs, probably related to the ability of the grafted liver to induce immunotolerance.
Collapse
Affiliation(s)
- Maria Rendina
- Gastroenterology Unit, University Hospital Policlinico of Bari, Bari, Italy
| | - Michele Barone
- Gastroenterology Unit, Department of Precision and Regenerative Medicine - Ionian Area-, University of Bari Aldo Moro, Bari, Italy
| | - Chiara Lillo
- Gastroenterology Unit, Department of Precision and Regenerative Medicine - Ionian Area-, University of Bari Aldo Moro, Bari, Italy
| | - Silvia Trapani
- Italian National Transplant Center, National Institute of Health, Rome, Italy
| | - Lucia Masiero
- Italian National Transplant Center, National Institute of Health, Rome, Italy
| | - Paolo Trerotoli
- Section of Statistics, Interdisciplinary Department of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Francesca Puoti
- Italian National Transplant Center, National Institute of Health, Rome, Italy
| | - Luigi Giovanni Lupo
- General Surgery and Liver Transplantation Unit, Department of Precision and Regenerative Medicine - Ionian Area-, University of Bari, Bari, Italy
| | - Francesco Tandoi
- General Surgery and Liver Transplantation Unit, Department of Precision and Regenerative Medicine - Ionian Area-, University of Bari, Bari, Italy
| | - Salvatore Agnes
- U.O.C. Chirurgia Generale e Trapianti di Organo, Policlinico Gemelli, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Antonio Grieco
- U.O.C. Medicina Interna e del Trapianto di Fegato, Policlinico Gemelli, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Enzo Andorno
- Chirurgia dei Trapianti di Fegato, IRCCS Ospedale Policlinico San Martino, University of Genoa, Genoa, Italy
| | - Simona Marenco
- Gastroenterology Unit, IRCCS Ospedale Policlinico San Martino, University of Genoa, Genoa, Italy
| | | | - Umberto Baccarani
- Centro Trapianto di Fegato, A.O.U.I. Udine, Università degli Studi di Udine, Udine, Italy
| | - Pierluigi Toniutto
- U.S.D. Epatologia e Trapianto di Fegato, A.O.U.I. Udine, Università degli Studi di Udine, Udine, Italy
| | | | - Antonio Colecchia
- Gastroenterology Unit, Department of Medical Specialties, University Hospital of Modena, Modena, Italy
| | - Matteo Cescon
- Chirurgia Epatobiliare e dei Trapianti, IRCCS, A.O.U. di Bologna, University of Bologna, Bologna, Italy
| | - Maria Cristina Morelli
- Internal Medicine Unit for the Treatment of Severe Organ Failure, IRCCS, A.O.U. di Bologna, Bologna, Italy
| | - Umberto Cillo
- Hepatobiliary Surgery and Liver Transplantation, University-Teaching Hospital of Padova, Padova, Italy
| | - Patrizia Burra
- Multivisceral Transplant Unit, University-Teaching Hospital of Padova, Padova, Italy
| | - Paolo Angeli
- Unit of Internal Medicine and Hepatology (UIMH), University-Teaching Hospital of Padova, Padova, Italy
| | - Michele Colledan
- U.O.C. Chirurgia Generale III, Centro Trapianti Fegato, A.S.S.T. Ospedale Papa Giovanni XXIII, Bergamo, Italy
| | - Stefano Fagiuoli
- Gastroenterology Hepatology and Transplantation Unit, A.S.S.T. Ospedale Papa Giovanni XXIII, Bergamo, Italy
- Gastroenterologia, Department of Medicine University of Milan Bicocca, Milano, Italy
| | - Luciano De Carlis
- Chirurgia Generale dei Trapianti, Azienda Ospedaliera Niguarda Ca’Granda, University of Milano-Bicocca, Milano, Italy
| | - Luca Belli
- U.O.C. Epatologia e Gastroenterologia, Azienda Ospedaliera Niguarda Ca’Granda, Milano, Italy
| | - Paolo De Simone
- U.O.C. Chirurgia Epatica e Trapianti di Fegato, A.O.U. Pisana, University of Pisa, Pisa, Italy
| | - Paola Carrai
- U.O. Chirurgia Epatica e del Trapianto di Fegato, A.O.U. Pisana, Pisa, Italy
| | - Fabrizio Di Benedetto
- U.O.C. di Chirurgia Oncologica Epatobiliopancreatica e Chirurgia dei Trapianti di Fegato, Azienda Ospedaliera Policlinico, Università di Modena, Modena, Italy
| | - Nicola De Maria
- U.O.C. Chirurgia Generale e Trapianti, Azienda Ospedaliera San Camillo Forlanini, Roma, Italy
| | | | - Valerio Giannelli
- Hepatology Unit, Azienda Ospedaliera San Camillo Forlanini, Roma, Italy
| | | | - Riccardo Volpes
- Unità di Gastroenterologia ed Epatologia, IRCCS-ISMETT-UPMCI, Palermo, Italy
| | - Sveva Corsale
- Unità di Gastroenterologia ed Epatologia, IRCCS-ISMETT-UPMCI, Palermo, Italy
| | - Vincenzo Mazzaferro
- Hepato-pancreatic-biliary surgery and Liver transplantation, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
- Department of Oncology, University of Milan, Milan, Italy
| | - Sherrie Bhoori
- Hepato-pancreatic-biliary surgery and Liver transplantation, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Renato Romagnoli
- Chirurgia Generale 2, Centro Trapianto Fegato A.O.U Città della Salute e della Scienza di Torino, Presidio Molinette, Torino, Italy
| | - Silvia Martini
- Gastroenteroly Unit, A.O.U Città della Salute e della Scienza di Torino, Presidio Molinette, Torino, Italy
| | - Giorgio Rossi
- Division of General and Liver Transplant Surgery, Ospedale Maggiore Policlinico, Milano, Italy
| | - Lucio Caccamo
- Division of General and Liver Transplant Surgery, Ospedale Maggiore Policlinico, Milano, Italy
| | - Maria Francesca Donato
- Division of Gastroenterology and Hepatology, Ospedale Maggiore Policlinico, Milano, Italy
| | - Massimo Rossi
- U.O.C. di Chirurgia Generale e Trapianti di Organo, Policlinico Umberto I, Sapienza Università di Roma, Rome, Italy
| | | | - Marco Spada
- Division of Hepatobiliopancreatic Surgery, Liver and Kidney Transplantation, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Giuseppe Maggiore
- Hepatogastroenterology, Digestive Endoscopy, Nutrition and Liver Transplantation Unit, Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Giuseppe Tisone
- Liver Transplant Unit, A.O.U. Policlinico Tor Vergata, University of Tor Vergata Rome, Rome, Italy
| | - Ilaria Lenci
- Hepatology Unit, A.O.U. Policlinico Tor Vergata, University of Tor Vergata Rome, Rome, Italy
| | - Giovanni Vennarecci
- Hepatobiliary and Liver Tranplantation Surgery, A.O.R.N. “A. CARDARELLI”, Naples, Italy
| | | | - Marco Vivarelli
- Chirurgia Epatobiliare, Pancreatica e dei Trapianti, A.O.U., Ospedali Riuniti, Ancona, Italy
| | | | - Fausto Zamboni
- General and Hepatic Transplantation Surgery Unit, AO.B. G. Brotzu, Cagliari, Italy
| | - Laura Mameli
- General and Hepatic Transplantation Surgery Unit, AO.B. G. Brotzu, Cagliari, Italy
| | - Silvio Tafuri
- Interdisciplinary Department of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Simona Simone
- Dialysis and Kidney Transplantation Unit, Department of Precision and Regenerative Medicine - Ionian Area-, University of Bari Aldo Moro, Bari, Italy
| | - Loreto Gesualdo
- Dialysis and Kidney Transplantation Unit, Department of Precision and Regenerative Medicine - Ionian Area-, University of Bari Aldo Moro, Bari, Italy
| | - Massimo Cardillo
- Italian National Transplant Center, National Institute of Health, Rome, Italy
| | - Alfredo Di Leo
- Gastroenterology Unit, Department of Precision and Regenerative Medicine - Ionian Area-, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
6
|
Citores MJ, Caballero-Marcos A, Cuervas-Mons V, Alonso-Fernández R, Graus-Morales J, Arias-Milla A, Valerio M, Muñoz P, Salcedo M. Long term SARS-CoV-2-specific cellular immunity after COVID-19 in liver transplant recipients. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2023:S1684-1182(23)00071-3. [PMID: 36964052 PMCID: PMC10020132 DOI: 10.1016/j.jmii.2023.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 02/09/2023] [Accepted: 03/07/2023] [Indexed: 03/19/2023]
Abstract
PURPOSE Long-term immunity after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in immunosuppressed patients is not well characterized. We aimed to explore the long-term natural immunity against SARS-CoV-2 in liver transplant (LT) recipients compared to the non-transplanted population (control group). METHODS Fifteen LT recipients and 15 controls matched according to variables associated with disease severity were included at 12 months following the coronavirus disease 2019 (COVID-19) onset. Peripheral blood mononuclear cells were stimulated with peptide pools covering spike (S), nucleocapside (N), and membrane (M) proteins. Reactive CD4+ and CD8+ T cells were identified using flow cytometry, and cytokine production was evaluated in the culture supernatants using cytometric bead array. Serum anti-N and anti-S IgG antibodies were detected with chemiluminescence. RESULTS The percentage of patients with a positive response in both CD4+ and CD8+ T cells against each viral protein and IL2, IL10, TNF-α, and IFN-γ levels was similar between LT recipients and controls. IFN-γ levels were positively correlated with the percentage of reactive CD4+ (p = 0.022) and CD8+ (p = 0.043) T cells to a mixture of M + N + S peptide pools. The prevalence and levels of anti-N and anti-S IgG antibodies were slightly lower in the LT recipients, but the difference was not statistically significant. CONCLUSION LT recipients exhibited a similar T cell response compared to non-transplanted individuals one year after COVID-19 diagnosis.
Collapse
Affiliation(s)
- Maria J Citores
- Laboratorio de Medicina Interna, Hospital Universitario Puerta de Hierro Majadahonda, IDIPHISA, Madrid, Spain.
| | - Aranzazu Caballero-Marcos
- Hepatology and Liver Transplantation Unit, Hospital General Universitario Gregorio Marañón, Facultad de Medicina Universidad Complutense, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain.
| | - Valentín Cuervas-Mons
- Unidad de Trasplante Hepático, Servicio de Medicina Interna, Hospital Universitario Puerta de Hierro Majadahonda, IDIPHISA, Madrid, Spain; Departamento de Medicina, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Madrid, Spain.
| | - Roberto Alonso-Fernández
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain.
| | - Javier Graus-Morales
- Department of Digestive Diseases, Hospital Ramón y Cajal, IRYCIS, Madrid, Spain.
| | - Ana Arias-Milla
- Unidad de Trasplante Hepático, Servicio de Medicina Interna, Hospital Universitario Puerta de Hierro Majadahonda, IDIPHISA, Madrid, Spain.
| | - Maricela Valerio
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain.
| | - Patricia Muñoz
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain.
| | - Magdalena Salcedo
- Hepatology and Liver Transplantation Unit, Hospital General Universitario Gregorio Marañón, Facultad de Medicina Universidad Complutense, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain.
| |
Collapse
|
7
|
Reeg DB, Hofmann M, Neumann-Haefelin C, Thimme R, Luxenburger H. SARS-CoV-2-Specific T Cell Responses in Immunocompromised Individuals with Cancer, HIV or Solid Organ Transplants. Pathogens 2023; 12:pathogens12020244. [PMID: 36839516 PMCID: PMC9966413 DOI: 10.3390/pathogens12020244] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Adaptive immune responses play an important role in the clinical course of SARS-CoV-2 infection. While evaluations of the virus-specific defense often focus on the humoral response, cellular immunity is crucial for the successful control of infection, with the early development of cytotoxic T cells being linked to efficient viral clearance. Vaccination against SARS-CoV-2 induces both CD4+ and CD8+ T cell responses and permits protection from severe COVID-19, including infection with the currently circulating variants of concern. Nevertheless, in immunocompromised individuals, first data imply significantly impaired SARS-CoV-2-specific immune responses after both natural infection and vaccination. Hence, these high-risk groups require particular consideration, not only in routine clinical practice, but also in the development of future vaccination strategies. In order to assist physicians in the guidance of immunocompromised patients, concerning the management of infection or the benefit of (booster) vaccinations, this review aims to provide a concise overview of the current knowledge about SARS-CoV-2-specific cellular immune responses in the vulnerable cohorts of cancer patients, people living with HIV (PLWH), and solid organ transplant recipients (SOT). Recent findings regarding the virus-specific cellular immunity in these differently immunocompromised populations might influence clinical decision-making in the future.
Collapse
|
8
|
Yang J, Lee KW, Baek JY, Bae S, Lee YH, Kim H, Huh K, Cho SY, Kang CI, Chung DR, Peck KR, Park JB, Kim SH, Kim TJ, Kim DM, Ko JH. Augmented humoral and cellular immunity against severe acute respiratory syndrome coronavirus 2 after breakthrough infection in kidney transplant recipients who received 3 doses of coronavirus disease 2019 vaccine. Am J Transplant 2023; 23:565-572. [PMID: 36739177 PMCID: PMC9807455 DOI: 10.1016/j.ajt.2022.12.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/21/2022] [Accepted: 12/17/2022] [Indexed: 01/04/2023]
Abstract
Diminished immune response to coronavirus disease 2019 (COVID-19) vaccines and breakthrough infection (BI) is a major concern for solid organ transplant recipients. Humoral and cellular immune responses of kidney transplant (KT) recipients after a third COVID-19 vaccination were investigated compared to matched health care workers. Anti-severe acute respiratory syndrome coronavirus 2 spike protein antibody and severe acute respiratory syndrome coronavirus 2 specific interferon-gamma releasing assay (IGRA) were assessed. A total of 38 KT recipients, including 20 BI and 18 noninfection, were evaluated. In the KT BI group, antibody titers were significantly increased (median 5 to 724, binding antibody units/mL (P = 0.002) after the third vaccination, but IGRA responses were negligible. After BI, antibody titers increased (median 11 355 binding antibody unit/mL; P < 0.001) and there was a significant increase of IGRA responses to spike proteins (Spike1-Nil, median 0.05 to 0.41 IU/mL; P = 0.009). Antibody titers and IGRA responses were significantly higher in the BI than in the noninfection group after 6 months. Immune responses were stronger in the health care worker than in the KT cohort, but the gap became narrower after BI. In conclusion, KT recipients who experienced BI after 3 COVID-19 vaccinations acquired augmented humoral and cellular immune responses.
Collapse
Affiliation(s)
- Jinyoung Yang
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyo Won Lee
- Division of Transplantation, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jin Yang Baek
- Asia Pacific Foundation for Infectious Diseases, Seoul, Republic of Korea
| | - Seongman Bae
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Young Ho Lee
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Haein Kim
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyungmin Huh
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sun Young Cho
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Cheol-In Kang
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Doo Ryeon Chung
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyong Ran Peck
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jae Berm Park
- Division of Transplantation, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sung-Han Kim
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Tae-Jong Kim
- Department of Rheumatology, Chonnam National University Medical School, Gwangju, South Korea
| | - Dong-Min Kim
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju, Republic of Korea.
| | - Jae-Hoon Ko
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Iryaningrum MR, Cahyadi A, Damara FA, Bandiara R, Marbun MBH. Seroconversion rates in kidney transplant recipients following SARS-CoV-2 vaccination and its association with immunosuppressive agents: a systematic review and meta-analysis. Clin Exp Vaccine Res 2023; 12:13-24. [PMID: 36844682 PMCID: PMC9950232 DOI: 10.7774/cevr.2023.12.1.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 12/27/2022] [Accepted: 12/27/2022] [Indexed: 02/19/2023] Open
Abstract
This systematic and meta-analysis aims to evaluate humoral and cellular responses to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine among kidney transplant recipients (KTRs). We conducted a systematic literature search across databases to evaluate seroconversion and cellular response rates in KTRs receiving SARS-CoV-2 vaccines. We extracted studies that assessed seroconversion rates described as the presence of antibody de novo positivity in KTRs following SARS-CoV-2 vaccination published up to January 23rd, 2022. We also performed meta-regression based on immunosuppression therapy used. A total of 44 studies involving 5,892 KTRs were included in this meta-analysis. The overall seroconversion rate following complete dose of vaccines was 39.2% (95% confidence interval [CI], 33.3%-45.3%) and cellular response rate was 41.6% (95% CI, 30.0%-53.6%). Meta-regression revealed that low antibody response rate was significantly associated with the high prevalence of mycophenolate mofetil/mycophenolic acid (p=0.04), belatacept (p=0.02), and anti-CD25 induction therapy uses (p=0.04). Conversely, tacrolimus use was associated with higher antibody response (p=0.01). This meta-analysis suggests that postvaccination seroconversion and cellular response rates in KTRs are still low. And seroconversion rate was correlated with the type of immunosuppressive agent and induction therapy used. Additional doses of the SARS-CoV-2 vaccine for this population using a different type of vaccine are considered.
Collapse
Affiliation(s)
- Maria Riastuti Iryaningrum
- Department of Internal Medicine, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Alius Cahyadi
- Department of Internal Medicine, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Fachreza Aryo Damara
- Dr Hasan Sadikin Hospital, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Ria Bandiara
- Department of Internal Medicine, Dr Hasan Sadikin Hospital, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Maruhum Bonar Hasiholan Marbun
- Department of Internal Medicine, Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
10
|
Fu J, Rust D, Fang Z, Jiao W, Lagana S, Batal I, Chen B, Merl S, Jones R, Sykes M, Weiner J. T cell repertoire profiling in allografts and native tissues in recipients with COVID-19 after solid organ transplantation: Insight into T cell-mediated allograft protection from viral infection. Front Immunol 2022; 13:1056703. [PMID: 36591281 PMCID: PMC9795050 DOI: 10.3389/fimmu.2022.1056703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/18/2022] [Indexed: 12/15/2022] Open
Abstract
Introduction The effects of the SARS-CoV-2 virus on the body, and why the effects are more severe in certain patients, remain incompletely understood. One population of special interest is transplant recipients because of their immunosuppressed state. Understanding the pathophysiology of graft dysfunction in transplant patients with the COVID-19 viral syndrome is important for prognosticating the risk to the graft as well as understanding how best to prevent and, if necessary, treat graft injury in these patients. Methods We analyzed multiple types of solid organ transplant recipients (liver, kidney, heart or lung) at our institution who died from SARS-CoV-2 and underwent autopsy (n = 6) or whose grafts were biopsied during active SARS-CoV-2 infection (n = 8). Their serum inflammatory markers were examined together with the histological appearance, viral load, and TCR repertoire of their graft tissue and, for autopsy patients, several native tissues. Results Histology and clinical lab results revealed a systemic inflammatory pattern that included elevated inflammatory markers and diffuse tissue damage regardless of graft rejection. Virus was detected throughout all tissues, although most abundant in lungs. The TCR repertoire was broadly similar throughout the tissues of each individual, with greater sharing of dominant clones associated with more rapid disease course. There was no difference in viral load or clonal distribution of overall, COVID-associated, or putative SARS-CoV-2-specific TCRs between allograft and native tissue. We further demonstrated that SARSCoV-2-specific TCR sequences in transplant patients lack a donor HLArestricted pattern, regardless of distribution in allograft or native tissues,suggesting that recognition of viral antigens on infiltrating recipient cells can effectively trigger host T cell anti-viral responses in both the host and graft. Discussion Our findings suggest a systemic immune response to the SARS-CoV-2 virus in solid organ transplant patients that is not associated with rejection and consistent with a largely destructive effect of recipient HLA-restricted T cell clones that affects donor and native organs similarly.
Collapse
Affiliation(s)
- Jianing Fu
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States,*Correspondence: Jianing Fu, ; Joshua Weiner,
| | - Dylan Rust
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Zhou Fang
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States
| | - Wenyu Jiao
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States
| | - Stephen Lagana
- Department of Pathology, Columbia University, New York, NY, United States
| | - Ibrahim Batal
- Department of Pathology, Columbia University, New York, NY, United States
| | - Bryan Chen
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States
| | - Sarah Merl
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States,Department of Pathology, Columbia University, New York, NY, United States
| | - Rebecca Jones
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States
| | - Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States,Department of Microbiology and Immunology, Columbia University, New York, NY, United States,Department of Surgery, Columbia University, New York, NY, United States
| | - Joshua Weiner
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States,Department of Surgery, Columbia University, New York, NY, United States,*Correspondence: Jianing Fu, ; Joshua Weiner,
| |
Collapse
|
11
|
Babel N, Hugo C, Westhoff TH. Vaccination in patients with kidney failure: lessons from COVID-19. Nat Rev Nephrol 2022; 18:708-723. [PMID: 35999285 PMCID: PMC9397175 DOI: 10.1038/s41581-022-00617-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2022] [Indexed: 02/06/2023]
Abstract
Infection is the second leading cause of death in patients with chronic kidney disease (CKD). Adequate humoral (antibody) and cellular (T cell-driven) immunity are required to minimize pathogen entry and promote pathogen clearance to enable infection control. Vaccination can generate cellular and humoral immunity against specific pathogens and is used to prevent many life-threatening infectious diseases. However, vaccination efficacy is diminished in patients with CKD. Premature ageing of the immune system and chronic systemic low-grade inflammation are the main causes of immune alteration in these patients. In the case of SARS-CoV-2 infection, COVID-19 can have considerable detrimental effects in patients with CKD, especially in those with kidney failure. COVID-19 prevention through successful vaccination is therefore paramount in this vulnerable population. Although patients receiving dialysis have seroconversion rates comparable to those of patients with normal kidney function, most kidney transplant recipients could not generate humoral immunity after two doses of the COVID-19 vaccine. Importantly, some patients who were not able to produce antibodies still had a detectable vaccine-specific T cell response, which might be sufficient to prevent severe COVID-19. Correlates of protection against SARS-CoV-2 have not been established for patients with kidney failure, but they are urgently needed to enable personalized vaccination regimens.
Collapse
Affiliation(s)
- Nina Babel
- Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany.
- Center for Translational Medicine and Immune Diagnostics Laboratory, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany.
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Center for Advanced Therapies (BeCAT) and Berlin Institute of Health, Berlin, Germany.
| | - Christian Hugo
- Medizinische Klinik und Poliklinik III, Universitätsklinikum, Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Timm H Westhoff
- Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| |
Collapse
|
12
|
Thümmler L, Konik M, Lindemann M, Fisenkci N, Koldehoff M, Gäckler A, Horn PA, Theodoropoulos F, Taube C, Zettler M, Anastasiou OE, Braß P, Jansen S, Witzke O, Rohn H, Krawczyk A. Long-term cellular immune response in immunocompromised unvaccinated COVID-19 patients undergoing monoclonal antibody treatment. Front Immunol 2022; 13:980698. [PMID: 36311723 PMCID: PMC9606643 DOI: 10.3389/fimmu.2022.980698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
Immunocompromised patients are at increased risk for a severe course of COVID-19. Treatment of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection with anti-SARS-CoV-2 monoclonal antibodies (mAbs) has become widely accepted. However, the effects of mAb treatment on the long-term primary cellular response to SARS-CoV-2 are unknown. In the following study, we investigated the long-term cellular immune responses to SARS-CoV-2 Spike S1, Membrane (M) and Nucleocapsid (N) antigens using the ELISpot assay in unvaccinated, mAb-treated immunocompromised high-risk patients. Anti-SARS-CoV-2 mAb untreated though vaccinated COVID-19 immunocompromised patients, vaccinated SARS-CoV-2 immunocompromised patients without COVID-19 and vaccinated healthy control subjects served as control groups. The cellular immune response was determined at a median of 5 months after SARS-CoV-2 infection. Our data suggest that immunocompromised patients develop an endogenous long-term cellular immune response after COVID-19, although at low levels. A better understanding of the cellular immune response will help guide clinical decision making for these vulnerable patient cohorts.
Collapse
Affiliation(s)
- Laura Thümmler
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Medicine Essen, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- Institute for Transfusion Medicine, University Medicine Essen, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Margarethe Konik
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Medicine Essen, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Monika Lindemann
- Institute for Transfusion Medicine, University Medicine Essen, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Neslinur Fisenkci
- Institute for Transfusion Medicine, University Medicine Essen, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Michael Koldehoff
- Department of Hematology and Stem Cell Transplantation, University Medicine Essen, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Hygiene and Environmental Medicine, University Medicine Essen, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Anja Gäckler
- Department of Nephrology, University Medicine Essen, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Peter A. Horn
- Institute for Transfusion Medicine, University Medicine Essen, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Fotis Theodoropoulos
- Department of Pneumology, University Medicine Essen-Ruhrlandklinik, University Duisburg- Essen, Essen, Germany
| | - Christian Taube
- Department of Pneumology, University Medicine Essen-Ruhrlandklinik, University Duisburg- Essen, Essen, Germany
| | - Markus Zettler
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Medicine Essen, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Olympia Evdoxia Anastasiou
- Institute for Virology, University Medicine Essen, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Peer Braß
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Medicine Essen, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Sarah Jansen
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Medicine Essen, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Oliver Witzke
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Medicine Essen, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Hana Rohn
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Medicine Essen, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- *Correspondence: Adalbert Krawczyk, ; Hana Rohn,
| | - Adalbert Krawczyk
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Medicine Essen, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- Institute for Virology, University Medicine Essen, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- *Correspondence: Adalbert Krawczyk, ; Hana Rohn,
| |
Collapse
|
13
|
Takiguchi S, Tomita Y, Uehara S, Tateishi K, Yamamoto N, Nakamura M. Immunological imprint on peripheral blood in kidney transplant recipients after two doses of SARS-CoV-2 mRNA vaccination in Japan. Front Med (Lausanne) 2022; 9:999374. [PMID: 36250073 PMCID: PMC9553995 DOI: 10.3389/fmed.2022.999374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
The immunological imprint after two doses of severe acute respiratory syndrome–coronavirus 2 (SARS-CoV-2) mRNA vaccination for patients after kidney transplantation (KTx) remain unclear. This study included KTx recipients and volunteer healthy controls (HCs) who received two doses of SARS-CoV-2 mRNA vaccine (Pfizer BioNTech) from January 2021 to December 2021. We analyzed safety within 21 days after each vaccination dose and compared the immune response in peripheral blood mononuclear cells (PBMCs) between the two groups. No graft rejection was observed throughout this study. Adverse events were generally observed within 5 days. The KTx group exhibited a significantly lower degree of symptoms between doses 1 and 2 (P < 0.001). Increases in activated subsets of T and B cells expressing human leukocyte antigen (HLA)-DR and/or CD38 were observed in the HC group after dose 2 (both P < 0.001), with the greatest increases in HLA-DR+CD8+ T cells and CD38+CD19+ B cells (P = 0.042 and P = 0.031, respectively). In addition, PD1+CD8+ T cells—but not PD1+CD4+ T cells—increased significantly in the HC group (P = 0.027). In the KTx group, however, activated HLA-DR+, CD38+, and PD1+ cells remained at baseline levels. Immunoglobulin (Ig)G against SARS-CoV-2 was detected in only four KTx recipients (13.3%) after dose 2 (P < 0.001). Multivariate logistic regression analyses revealed that ΔHLA-DR+CD8+ T cells and ΔCD38+CD19+ B cells were significantly associated with IgG formation (both P = 0.02). SARS-CoV-2 mRNA vaccine generates impaired cellular and humoral immunity for KTx recipients. Results indicate the need for modified vaccination strategies in immunocompromised KTx recipients.
Collapse
Affiliation(s)
- Shinya Takiguchi
- Department of Transplant Surgery, Tokai University School of Medicine, Kanagawa, Japan
| | - Yusuke Tomita
- Department of Transplant Surgery, Tokai University School of Medicine, Kanagawa, Japan
- *Correspondence: Yusuke Tomita, ; orcid.org/0000-0003-4959-6290
| | - Saeko Uehara
- Department of Transplant Surgery, Tokai University School of Medicine, Kanagawa, Japan
| | - Koichiro Tateishi
- Department of Virology, Division of Host Defense Mechanism, School of Medicine, Tokai University, Kanagawa, Japan
| | - Norio Yamamoto
- Department of Virology, Division of Host Defense Mechanism, School of Medicine, Tokai University, Kanagawa, Japan
| | - Michio Nakamura
- Department of Transplant Surgery, Tokai University School of Medicine, Kanagawa, Japan
| |
Collapse
|
14
|
Opsomer R, Kuypers D. COVID-19 and solid organ transplantation: Finding the right balance. Transplant Rev (Orlando) 2022; 36:100710. [PMID: 35809422 PMCID: PMC9251959 DOI: 10.1016/j.trre.2022.100710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Roxanne Opsomer
- Catholic University of Leuven, Faculty of Medicine, Herestraat 49, 3000 Leuven, Belgium.
| | - Dirk Kuypers
- University Hospitals Leuven, Department of Nephrology and Renal Transplantation; Catholic University Leuven, Department of Microbiology, Immunology and Transplantation, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
15
|
Rahmani A, Dini G, Leso V, Montecucco A, Kusznir Vitturi B, Iavicoli I, Durando P. Duration of SARS-CoV-2 shedding and infectivity in the working age population: a systematic review and meta-analysis. LA MEDICINA DEL LAVORO 2022; 113:e2022014. [PMID: 35481581 PMCID: PMC9073762 DOI: 10.23749/mdl.v113i2.12724] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/22/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND During the COVID-19 pandemic, working age individuals have been implicated in sustaining the resurgence of SARS-CoV-2 infections, and multiple outbreaks have been observed in several occupational settings. In this regard, Occupational Physicians play a crucial role in the management of infected workers, particularly in the safe return-to-work of subjects after clinical resolution. To this end, knowledge of the duration of the infective phase in the working age population is essential, taking into account previous evidence suggesting that PCR positivity does not coincide with virus viability. METHODS A systematic review and meta-analysis, searching major scientific databases, including PubMed/MEDLINE, Scopus and Web of Science, were performed in order to synthesize the available evidence regarding the mean and maximal duration of infectivity compared to the mean and maximal duration of viral RNA shedding. A subgroup analysis of the studies was performed according to the immunocompetent or immunocompromised immune status of the majority of the enrolled individuals. RESULTS Twenty studies were included in the final qualitative and quantitative analysis (866 individuals). Overall, a mean duration of RT-PCR positivity after symptom onset was found equal to 27.9 days (95%CI 23.3-32.5), while the mean duration of replicant competent virus isolation was 7.3 days (95%CI 5.7-8.8). The mean duration of SARS-CoV-2 shedding resulted equal to 26.5 days (95%CI 21.4-31.6) and 36.3 days (95%CI 21.9-50.6), and the mean duration of SARS-CoV-2 infectivity was 6.3 days (95%CI 4.9-7.8) and 29.5 days (95%CI 12.5-46.5), respectively considering immunocompetent and immunocompromised individuals. The maximum duration of infectivity among immunocompetent subjects was reported after 18 days from symptom onset, while in immunocompromised individuals it lasted up to 112 days. CONCLUSIONS These findings suggest that the test-based strategy before return-to-work might not be warranted after 21 days among immunocompetent working age individuals, and could keep many workers out of occupation, reducing their livelihood and productivity.
Collapse
Affiliation(s)
- Alborz Rahmani
- Department of Health Sciences, University of Genoa; Occupational Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Guglielmo Dini
- Department of Health Sciences, University of Genoa; Occupational Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Veruscka Leso
- Section of Occupational Medicine, Department of Public Health, University of Naples Federico II, Naples, Italy.
| | - Alfredo Montecucco
- Department of Health Sciences, University of Genoa; Occupational Medicine Unit, IRCCS Ospedale Policlinico San Martino.
| | | | - Ivo Iavicoli
- Section of Occupational Medicine, Department of Public Health, University of Naples Federico II, Naples, Italy.
| | - Paolo Durando
- Department of Health Sciences, University of Genoa; Occupational Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| |
Collapse
|
16
|
Sanders JSF, Bemelman FJ, Messchendorp AL, Baan CC, van Baarle D, van Binnendijk R, Diavatopoulos DA, Frölke SC, Geers D, GeurtsvanKessel CH, den Hartog G, van der Heiden M, Imhof C, Kho MM, Koopmans MP, Malahe SRK, Mattheussens WB, van der Molen R, van Mourik D, Remmerswaal EB, Rots N, Vart P, de Vries RD, Gansevoort RT, Hilbrands LB, Reinders ME. The RECOVAC Immune-response Study: The Immunogenicity, Tolerability, and Safety of COVID-19 Vaccination in Patients With Chronic Kidney Disease, on Dialysis, or Living With a Kidney Transplant. Transplantation 2022; 106:821-834. [PMID: 34753894 PMCID: PMC8942603 DOI: 10.1097/tp.0000000000003983] [Citation(s) in RCA: 123] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 09/22/2021] [Accepted: 09/26/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND In kidney patients COVID-19 is associated with severely increased morbidity and mortality. A comprehensive comparison of the immunogenicity, tolerability, and safety of COVID-19 vaccination in different cohorts of kidney patients and a control cohort is lacking. METHODS This investigator driven, prospective, controlled multicenter study included 162 participants with chronic kidney disease (CKD) stages G4/5 (eGFR < 30 mL/min/1.73m2), 159 participants on dialysis, 288 kidney transplant recipients, and 191 controls. Participants received 2 doses of the mRNA-1273 COVID-19 vaccine (Moderna). The primary endpoint was seroconversion. RESULTS Transplant recipients had a significantly lower seroconversion rate when compared with controls (56.9% versus 100%, P < 0.001), with especially mycophenolic acid, but also, higher age, lower lymphocyte concentration, lower eGFR, and shorter time after transplantation being associated with nonresponder state. Transplant recipients also showed significantly lower titers of neutralizing antibodies and T-cell responses when compared with controls. Although a high seroconversion rate was observed for participants with CKD G4/5 (100%) and on dialysis (99.4%), mean antibody concentrations in the CKD G4/5 cohort and dialysis cohort were lower than in controls (2405 [interquartile interval 1287-4524] and 1650 [698-3024] versus 3186 [1896-4911] BAU/mL, P = 0.06 and P < 0.001, respectively). Dialysis patients and especially kidney transplant recipients experienced less systemic vaccination related adverse events. No specific safety issues were noted. CONCLUSIONS The immune response following vaccination in patients with CKD G4/5 and on dialysis is almost comparable to controls. In contrast, kidney transplant recipients have a poor response. In this latter, patient group development of alternative vaccination strategies are warranted.
Collapse
Affiliation(s)
- Jan-Stephan F. Sanders
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Frederike J. Bemelman
- Renal Transplant Unit, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - A. Lianne Messchendorp
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Carla C. Baan
- Department of Internal Medicine, Nephrology, and Transplantation, Erasmus MC Transplant Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Debbie van Baarle
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, Groningen, The Netherlands
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Rob van Binnendijk
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Dimitri A. Diavatopoulos
- Radboud Center for Infectious Diseases, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands
| | - Sophie C. Frölke
- Renal Transplant Unit, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Daryl Geers
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Gerco den Hartog
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Marieke van der Heiden
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, Groningen, The Netherlands
| | - Celine Imhof
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, Groningen, The Netherlands
| | - Marcia M.L. Kho
- Department of Internal Medicine, Nephrology, and Transplantation, Erasmus MC Transplant Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | - Wouter B. Mattheussens
- Department of Nephrology, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Renate van der Molen
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands
| | - Djenolan van Mourik
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ester B.M. Remmerswaal
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Nynke Rots
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Priya Vart
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rory D. de Vries
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ron T. Gansevoort
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Luuk B. Hilbrands
- Department of Nephrology, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Marlies E.J. Reinders
- Department of Internal Medicine, Nephrology, and Transplantation, Erasmus MC Transplant Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - RECOVAC Collaborators*
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Renal Transplant Unit, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Internal Medicine, Nephrology, and Transplantation, Erasmus MC Transplant Institute, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, Groningen, The Netherlands
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Nephrology, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
17
|
Wünsch K, Anastasiou OE, Alt M, Brochhagen L, Cherneha M, Thümmler L, van Baal L, Madel RJ, Lindemann M, Taube C, Witzke O, Rohn H, Krawczyk A, Jansen S. COVID-19 in Elderly, Immunocompromised or Diabetic Patients—From Immune Monitoring to Clinical Management in the Hospital. Viruses 2022; 14:v14040746. [PMID: 35458476 PMCID: PMC9024512 DOI: 10.3390/v14040746] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/19/2022] [Accepted: 03/21/2022] [Indexed: 01/01/2023] Open
Abstract
The novel, highly transmissible severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has triggered a pandemic of acute respiratory illness worldwide and remains a huge threat to the healthcare system’s capacity to respond to COVID-19. Elderly and immunocompromised patients are at increased risk for a severe course of COVID-19. These high-risk groups have been identified as developing diminished humoral and cellular immune responses. Notably, SARS-CoV-2 RNA remains detectable in nasopharyngeal swabs of these patients for a prolonged period of time. These factors complicate the clinical management of these vulnerable patient groups. To date, there are no well-defined guidelines for an appropriate duration of isolation for elderly and immunocompromised patients, especially in hospitals or nursing homes. The aim of the present study was to characterize at-risk patient cohorts capable of producing a replication-competent virus over an extended period after symptomatic COVID-19, and to investigate the humoral and cellular immune responses and infectivity to provide a better basis for future clinical management. In our cohort, the rate of positive viral cultures and the sensitivity of SARS-CoV-2 antigen tests correlated with higher viral loads. Elderly patients and patients with diabetes mellitus had adequate cellular and humoral immune responses to SARS-CoV-2 infection, while immunocompromised patients had reduced humoral and cellular immune responses. Our patient cohort was hospitalized for longer compared with previously published cohorts. Longer hospitalization was associated with a high number of nosocomial infections, representing a potential hazard for additional complications to patients. Most importantly, regardless of positive SARS-CoV-2 RNA detection, no virus was culturable beyond a cycle threshold (ct) value of 33 in the majority of samples. Our data clearly indicate that elderly and diabetic patients develop a robust immune response to SARS-CoV-2 and may be safely de-isolated at a ct value of more than 35.
Collapse
Affiliation(s)
- Korbinian Wünsch
- West German Centre of Infectious Diseases, Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (K.W.); (M.A.); (L.B.); (M.C.); (L.T.); (R.J.M.); (O.W.); (A.K.)
| | - Olympia E. Anastasiou
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany;
| | - Mira Alt
- West German Centre of Infectious Diseases, Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (K.W.); (M.A.); (L.B.); (M.C.); (L.T.); (R.J.M.); (O.W.); (A.K.)
| | - Leonie Brochhagen
- West German Centre of Infectious Diseases, Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (K.W.); (M.A.); (L.B.); (M.C.); (L.T.); (R.J.M.); (O.W.); (A.K.)
| | - Maxim Cherneha
- West German Centre of Infectious Diseases, Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (K.W.); (M.A.); (L.B.); (M.C.); (L.T.); (R.J.M.); (O.W.); (A.K.)
| | - Laura Thümmler
- West German Centre of Infectious Diseases, Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (K.W.); (M.A.); (L.B.); (M.C.); (L.T.); (R.J.M.); (O.W.); (A.K.)
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany;
| | - Lukas van Baal
- Department of Endocrinology, Diabetes and Metabolism and Division of Laboratory Research, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany;
| | - Rabea J. Madel
- West German Centre of Infectious Diseases, Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (K.W.); (M.A.); (L.B.); (M.C.); (L.T.); (R.J.M.); (O.W.); (A.K.)
| | - Monika Lindemann
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany;
| | - Christian Taube
- Department of Pneumology, University Medicine Essen—Ruhrlandklinik, University Duisburg-Essen, 45147 Essen, Germany;
| | - Oliver Witzke
- West German Centre of Infectious Diseases, Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (K.W.); (M.A.); (L.B.); (M.C.); (L.T.); (R.J.M.); (O.W.); (A.K.)
| | - Hana Rohn
- West German Centre of Infectious Diseases, Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (K.W.); (M.A.); (L.B.); (M.C.); (L.T.); (R.J.M.); (O.W.); (A.K.)
- Correspondence: (H.R.); (S.J.)
| | - Adalbert Krawczyk
- West German Centre of Infectious Diseases, Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (K.W.); (M.A.); (L.B.); (M.C.); (L.T.); (R.J.M.); (O.W.); (A.K.)
| | - Sarah Jansen
- West German Centre of Infectious Diseases, Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (K.W.); (M.A.); (L.B.); (M.C.); (L.T.); (R.J.M.); (O.W.); (A.K.)
- Correspondence: (H.R.); (S.J.)
| |
Collapse
|
18
|
Reischig T, Kacer M, Vlas T, Drenko P, Kielberger L, Machova J, Topolcan O, Kucera R, Kormunda S. Insufficient response to mRNA SARS-CoV-2 vaccine and high incidence of severe COVID-19 in kidney transplant recipients during pandemic. Am J Transplant 2022; 22:801-812. [PMID: 34860470 PMCID: PMC9906453 DOI: 10.1111/ajt.16902] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 01/25/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mRNA vaccination may fail to sufficiently protect transplant recipients against coronavirus disease 2019 (COVID-19). We retrospectively evaluated COVID-19 in kidney transplant recipients (n = 226) after BNT162b2 mRNA vaccine administration. The control group consisted of unvaccinated patients (n = 194) during the previous pandemic wave. We measured anti-spike protein immunoglobulin G (IgG) levels and cellular responses, using enzyme-linked immunosorbent spot assay, in a prospective cohort after vaccination (n = 31) and recovery from COVID-19 (n = 19). COVID-19 was diagnosed in 37 (16%) vaccinated and 43 (22%) unvaccinated patients. COVID-19 severity was similar in both groups, with patients exhibiting a comparable need for hospitalization (41% vs. 40%, p = 1.000) and mortality (14% vs. 9%, p = .726). Short posttransplant periods were associated with COVID-19 after vaccination (p < .001). Only 5 (16%) patients achieved positive SARS-CoV-2 IgG after vaccination, and 17 (89%, p < .001) recovered from COVID-19 (median IgG levels, 0.6 vs. 52.5 AU/ml, p < .001). A cellular response following vaccination was present in the majority (n = 22, 71%), with an increase in interleukin 2 secreting T cells (p < .001). Despite detectable T cell immunity after mRNA vaccination, kidney transplant recipients remained at a high risk of severe COVID-19. Humoral responses induced by vaccination were significantly lower than that after COVID-19.
Collapse
Affiliation(s)
- Tomas Reischig
- Department of Internal Medicine I, Faculty of Medicine in Pilsen, Charles University and Teaching Hospital, Pilsen, Czech Republic
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Martin Kacer
- Department of Internal Medicine I, Faculty of Medicine in Pilsen, Charles University and Teaching Hospital, Pilsen, Czech Republic
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Tomas Vlas
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
- Department of Immunology and Allergology, Faculty of Medicine in Pilsen, Charles University and Teaching Hospital, Pilsen, Czech Republic
| | - Petr Drenko
- Department of Internal Medicine I, Faculty of Medicine in Pilsen, Charles University and Teaching Hospital, Pilsen, Czech Republic
| | - Lukas Kielberger
- Department of Internal Medicine I, Faculty of Medicine in Pilsen, Charles University and Teaching Hospital, Pilsen, Czech Republic
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Jana Machova
- Department of Internal Medicine I, Faculty of Medicine in Pilsen, Charles University and Teaching Hospital, Pilsen, Czech Republic
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Ondrej Topolcan
- Department of Immunochemistry Diagnostics, Faculty of Medicine in Pilsen, Charles University and Teaching Hospital, Pilsen, Czech Republic
| | - Radek Kucera
- Department of Immunochemistry Diagnostics, Faculty of Medicine in Pilsen, Charles University and Teaching Hospital, Pilsen, Czech Republic
| | - Stanislav Kormunda
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
- Division of Information Technologies and Statistics, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| |
Collapse
|
19
|
SARS-CoV-2 cross-reactive B and T cell responses in kidney-transplant patients. Transplant Proc 2022; 54:1455-1464. [PMID: 35489983 PMCID: PMC8923882 DOI: 10.1016/j.transproceed.2022.02.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/18/2022] [Accepted: 02/28/2022] [Indexed: 11/19/2022]
Abstract
Background Immune responses to seasonal endemic coronaviruses might have a pivotal role in protection against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Those SARS-CoV-2-crossreactive T cells were recently described in immunocompetent individuals. Still, data on cross-reactive humoral and cellular immunity in kidney transplant recipients is currently lacking. Methods The pre-existing, cross-reactive antibody B and T cell immune responses against SARS-CoV-2 in unexposed adults with kidney transplantation (Tx, n = 14) and without (non-Tx, n = 12) sampled before the pandemic were compared with 22 convalescent patients with COVID-19 (Cp) applying enzyme-linked immunosorbent assay and flow cytometry. Results In both unexposed groups, SARS-CoV-2 IgG antibodies were not detectable. Memory B cells binding spike (S) protein SARS-CoV-2 were detected in unexposed individuals (64% among Tx; 50% among non-Tx) and higher frequencies after infection (80% Cp). The numbers of SARS-CoV-2-reactive T cells were comparable between patients who had undergone Tx and those who had not. SARS-CoV-2-reactive follicular T helper cells were present in 61% of the unexposed cohort in both patients who had undergone Tx and those who had not. Conclusions Cross-reactive memory B and T cells against SARS-CoV-2 exist also in transplanted adults, suggesting a primed adaptive immunity. The effect on the disease course may depend on the concomitant immunosuppressive drugs.
Collapse
|
20
|
Favà A, Donadeu L, Jouve T, Gonzalez-Costello J, Lladó L, Santana C, Toapanta N, Lopez M, Pernin V, Facundo C, Cabañas NS, Thaunat O, Crespo M, Llinàs-Mallol L, Revuelta I, Sabé N, Rombauts A, Calatayud L, Ardanuy C, Esperalba J, Fernandez C, Lozano JJ, Preyer R, Strecker K, Couceiro C, García-Romero E, Cachero A, Meneghini M, Torija A, Le Quintrec M, Melilli E, Cruzado JM, Polo C, Moreso F, Crespo E, Bestard O. A comprehensive assessment of long-term SARS-CoV-2-specific adaptive immune memory in convalescent COVID-19 Solid Organ Transplant recipients. Kidney Int 2022; 101:1027-1038. [PMID: 35124011 PMCID: PMC8813192 DOI: 10.1016/j.kint.2021.12.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 11/25/2021] [Accepted: 12/09/2021] [Indexed: 01/08/2023]
Affiliation(s)
- Alexandre Favà
- Kidney Transplant Unit, Nephrology Department, Bellvitge University Hospital, Barcelona, Spain
| | - Laura Donadeu
- Laboratory of Nephrology and Transplantation, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Thomas Jouve
- Laboratory of Nephrology and Transplantation, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Laura Lladó
- Liver Transplant Unit, Bellvitge University Hospital, Barcelona, Spain
| | - Carolina Santana
- Primary Care Baix Llobregat Centre, Direcció d'Atenció Primària Costa Ponent, Equip d'Atenció Primària Gavarra, Cornellà de Llobregat, Barcelona, Spain
| | - Néstor Toapanta
- Kidney Transplant Unit, Nephrology Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Manuel Lopez
- Lung Transplant Unit, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Vincent Pernin
- Kidney Transplant Unit, Hospital de Montpellier, Montpellier, France
| | - Carme Facundo
- Kidney Transplant Unit, Nephrology Department, Fundació PuigVert, Barcelona, Spain
| | - Nuria Serra Cabañas
- Kidney Transplant Unit, Nephrology Department, Fundació PuigVert, Barcelona, Spain
| | - Olivier Thaunat
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital Lyon, Hospices Civils de Lyon, Lyon, France
| | - Marta Crespo
- Kidney Transplant Unit, Nephrology Department, Hospital del Mar, Barcelona, Spain
| | - Laura Llinàs-Mallol
- Kidney Transplant Unit, Nephrology Department, Hospital del Mar, Barcelona, Spain
| | - Ignacio Revuelta
- Kidney Transplant Unit, Nephrology Department, Hospital Clínic, Barcelona, Spain
| | - Nuria Sabé
- Infectious Disease Department, Bellvitge University Hospital, Barcelona, Spain
| | - Alexander Rombauts
- Infectious Disease Department, Bellvitge University Hospital, Barcelona, Spain
| | - Laura Calatayud
- Microbiology Department, Bellvitge University Hospital, Barcelona, Spain
| | - Carmen Ardanuy
- Microbiology Department, Bellvitge University Hospital, Barcelona, Spain
| | - Juliana Esperalba
- Microbiology Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Candela Fernandez
- Microbiology Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Juan J Lozano
- Bioinformatics Platform, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Catalonia, Spain
| | | | | | - Carlos Couceiro
- Kidney Transplant Unit, Nephrology Department, Bellvitge University Hospital, Barcelona, Spain
| | | | - Alba Cachero
- Liver Transplant Unit, Bellvitge University Hospital, Barcelona, Spain
| | - Maria Meneghini
- Kidney Transplant Unit, Nephrology Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alba Torija
- Laboratory of Nephrology and Transplantation, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Edoardo Melilli
- Kidney Transplant Unit, Nephrology Department, Bellvitge University Hospital, Barcelona, Spain
| | - Josep Maria Cruzado
- Kidney Transplant Unit, Nephrology Department, Bellvitge University Hospital, Barcelona, Spain
| | - Carolina Polo
- Kidney Transplant Unit, Nephrology Department, Bellvitge University Hospital, Barcelona, Spain
| | - Francesc Moreso
- Kidney Transplant Unit, Nephrology Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Elena Crespo
- Laboratory of Nephrology and Transplantation, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Oriol Bestard
- Kidney Transplant Unit, Nephrology Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
21
|
Yelin D, Rozen-Zvi B, Yahav D, Ben-Dor N, Steinmetz T, Agur T, Zingerman B, Schneider S, Lichtenberg S, Ben-Zvi H, Mashraki T, Rahamimov R. OUP accepted manuscript. Clin Kidney J 2022; 15:992-998. [PMID: 35498878 PMCID: PMC8903319 DOI: 10.1093/ckj/sfac031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Indexed: 11/29/2022] Open
Abstract
Data regarding immunogenicity of mRNA severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines among kidney transplant recipients in the months following vaccination are lacking. We aimed to investigate humoral immune response at 3–4 months post-vaccination among a cohort of kidney transplant recipients, compared with a control group of dialysis patients. Anti-spike antibodies were tested at 1 and 3–4 months after vaccination. Of 259 kidney transplant recipients tested at a median time of 110 days from second vaccine dose, 99 (38%) were seropositive, compared with 83% (101/122) of control patients. Younger age, better renal function and lower immunosuppression levels were associated with seropositivity. A total of 14% (13/94) of participants seropositive at 1 month became seronegative at follow-up and 11% (18/165) became seropositive. The latter were mainly individuals with higher antibody levels at 1 month. Antibody levels at 3–4 months were significantly reduced in both study groups, although the decline was more pronounced in the control group. Kidney transplant recipients present poor antibody response to mRNA SARS-CoV-2 vaccination, with only 38% seropositive at 3–4 months. Nevertheless, the decay in antibody response over time is modest, and some patients may present delayed response, reaching adequate antibody levels at 3–4 months. Low seropositivity rates in this group call for investigating other immunization strategies.
Collapse
Affiliation(s)
| | | | - Dafna Yahav
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Infectious Diseases Unit, Rabin Medical Center, Beilinson Campus, Petah-Tikva, Israel
| | - Naomi Ben-Dor
- Department of Nephrology and Hypertension, Rabin Medical Center, Beilinson Campus, Petah-Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tali Steinmetz
- Department of Nephrology and Hypertension, Rabin Medical Center, Beilinson Campus, Petah-Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Timna Agur
- Department of Nephrology and Hypertension, Rabin Medical Center, Beilinson Campus, Petah-Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Boris Zingerman
- Department of Nephrology and Hypertension, Rabin Medical Center, Beilinson Campus, Petah-Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shira Schneider
- Department of Nephrology and Hypertension, Rabin Medical Center, Beilinson Campus, Petah-Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shelly Lichtenberg
- Department of Nephrology and Hypertension, Rabin Medical Center, Beilinson Campus, Petah-Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Haim Ben-Zvi
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Clinical Microbiology Laboratory, Rabin Medical Center, Beilinson Hospital, Petah-Tikva, Israel
| | - Tiki Mashraki
- Department of Nephrology and Hypertension, Rabin Medical Center, Beilinson Campus, Petah-Tikva, Israel
- Department of Transplantation, Rabin Medical Center, Beilinson Campus, Petah-Tikva, Israel
| | - Ruth Rahamimov
- Department of Nephrology and Hypertension, Rabin Medical Center, Beilinson Campus, Petah-Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Transplantation, Rabin Medical Center, Beilinson Campus, Petah-Tikva, Israel
| |
Collapse
|
22
|
Kute V, Desai S, Meshram H, Shah K, Chauhan S, Mishra V, Sahay M. On the way to mandate booster vaccine dose of coronavirus disease 2019 for transplant recipients: A narrative review of literature. INDIAN JOURNAL OF TRANSPLANTATION 2022. [DOI: 10.4103/ijot.ijot_7_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
23
|
Immune Response after SARS-CoV-2 Vaccination in Kidney Transplant Patients. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:medicina57121327. [PMID: 34946272 PMCID: PMC8703670 DOI: 10.3390/medicina57121327] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/22/2021] [Accepted: 12/01/2021] [Indexed: 01/14/2023]
Abstract
Background and Objectives: The prospective study was conducted to evaluate humoral and cellular immune responses after two doses of BNT162b2 (Pfizer-BioNTech) vaccine and possible relation with other factors (medication, etc.) in kidney transplant patients. Materials and Methods: Out of 167 vaccinated patients, 136 agreed to a follow-up visit three to six weeks after vaccination. Results: Only 39 patients (29%) developed antibody response against SARS-CoV-2 (≥35.2 binding antibody units (BAU)/mL) after full vaccination. Multivariate binary logistic regression analysis showed that predictive factors for good antibody response to the COVID-19 vaccine were better kidney function, higher hemoglobin level, and no use of mycophenolate mofetil for immunosuppression. For seropositive kidney transplant patients there was a significant negative correlation between anti-SARS-CoV-2 antibody titer and CD4/CD8 ratio (Spearman's correlation coefficient -0.4, p = 0.02), percentage of CD19+ cells (r = -0.37, p = 0.02), and a positive correlation with percentage of CD8+ cells (r = 0.4, p = 0.01). There was an increase of total leucocyte count after vaccination in the total studied population, and in the group of responders. Conclusions: Only one third of kidney transplant patients develop sufficient antibody responses after full COVID-19 vaccination with Pfizer-BioNTech. Better kidney function, higher hemoglobin level, and no use of mycophenolate mofetil for immunosuppression increases the adequacy of response. The antibody titers correlated positively with relative number of CD8+ cells and negatively with CD4/CD8 ratio in responders.
Collapse
|
24
|
Malipiero G, Moratto A, Infantino M, D'Agaro P, Piscianz E, Manfredi M, Grossi V, Benvenuti E, Bulgaresi M, Benucci M, Villalta D. Assessment of humoral and cellular immunity induced by the BNT162b2 SARS-CoV-2 vaccine in healthcare workers, elderly people, and immunosuppressed patients with autoimmune disease. Immunol Res 2021; 69:576-583. [PMID: 34417958 PMCID: PMC8379062 DOI: 10.1007/s12026-021-09226-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 08/11/2021] [Indexed: 01/01/2023]
Abstract
The development of vaccines to prevent SARS-CoV-2 infection has mainly relied on the induction of neutralizing antibodies (nAbs) to the Spike protein of SARS-CoV-2, but there is growing evidence that T cell immune response can contribute to protection as well. In this study, an anti-receptor binding domain (RBD) antibody assay and an INFγ-release assay (IGRA) were used to detect humoral and cellular responses to the Pfizer-BioNTech BNT162b2 vaccine in three separate cohorts of COVID-19-naïve patients: 108 healthcare workers (HCWs), 15 elderly people, and 5 autoimmune patients treated with immunosuppressive agents. After the second dose of vaccine, the mean values of anti-RBD antibodies (Abs) and INFγ were 123.33 U/mL (range 27.55-464) and 1513 mIU/mL (range 145-2500) in HCWs and 210.7 U/mL (range 3-500) and 1167 mIU/mL (range 83-2500) in elderly people. No correlations between age and immune status were observed. On the contrary, a weak but significant positive correlation was found between INFγ and anti-RBD Abs values (rho = 0.354, p = 0.003). As to the autoimmune cohort, anti-RBD Abs were not detected in the two patients with absent peripheral CD19+B cells, despite high INFγ levels being observed in all 5 patients after vaccination. Even though the clinical relevance of T cell response has not yet been established as a correlate of vaccine-induced protection, IGRA testing has showed optimal sensitivity and specificity to define vaccine responders, even in patients lacking a cognate antibody response to the vaccine.
Collapse
Affiliation(s)
- Giacomo Malipiero
- Immunology and Allergy Unit, Ospedale S-Maria degli Angeli, Pordenone, Italy
| | - Anna Moratto
- Immunology and Allergy Unit, Ospedale S-Maria degli Angeli, Pordenone, Italy
| | - Maria Infantino
- Immunology and Allergy Laboratory Unit, San Giovanni Di Dio Hospital, Florence, Italy
| | - Pierlanfranco D'Agaro
- Laboratory for Hygiene and Public Health, University Hospital of Trieste, Trieste, Italy
| | - Elisa Piscianz
- Laboratory for Hygiene and Public Health, University Hospital of Trieste, Trieste, Italy
| | - Mariangela Manfredi
- Immunology and Allergy Laboratory Unit, San Giovanni Di Dio Hospital, Florence, Italy
| | - Valentina Grossi
- Immunology and Allergy Laboratory Unit, San Giovanni Di Dio Hospital, Florence, Italy
| | - Enrico Benvenuti
- Geriatric Unit Firenze-Empoli, Santa Maria Annunziata Hospital, Florence, Italy
| | - Matteo Bulgaresi
- Geriatric Unit Firenze-Empoli, Santa Maria Annunziata Hospital, Florence, Italy
| | - Maurizio Benucci
- Rheumatology Unit, San Giovanni Di Dio Hospital, Florence, Italy
| | - Danilo Villalta
- Immunology and Allergy Unit, Ospedale S-Maria degli Angeli, Pordenone, Italy.
| |
Collapse
|
25
|
Bertrand D, Hamzaoui M, Drouot L, Lamulle J, Hanoy M, Edet S, Laurent C, Lebourg L, Etienne I, Lemoine M, Le Roy F, Nezam D, Mauger E, Boyer O, Guerrot D, Candon S. SARS-CoV-2-specific Humoral and Cellular Immunities in Kidney Transplant Recipients and Dialyzed Patients Recovered From Severe and Nonsevere COVID-19. Transplant Direct 2021; 7:e792. [PMID: 34805494 PMCID: PMC8601300 DOI: 10.1097/txd.0000000000001230] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/16/2021] [Accepted: 07/17/2021] [Indexed: 12/23/2022] Open
Abstract
Kidney transplantation and dialysis are two major risk factors for severe forms of coronavirus disease 2019 (COVID-19). The dynamics of the immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in this population remain largely unknown. Methods We report here the analysis of anti-SARS-CoV-2 antibody- and T cell-mediated immune responses in 26 kidney transplant recipients (KTRs) and 11 dialyzed patients (DPs) who recovered from COVID-19. Results After a mean time of 83 ± 26 d post-symptom onset for KTRs and 97 ± 31 d for DPs, 20 KTRs (76.9%) and 10 DPs (90.9%) displayed anti-S1 immunoglobulin G SARS-CoV-2 antibodies (P = 0.34), at similar titers in both groups. SARS-CoV-2-specific interferon-γ-producing T cells were evidenced in 26 KTRs (100%) and 10 DPs (90.9%). Total numbers of SARS-CoV-2-reactive T cells were high and not statistically different between the 2 groups. No correlation between the severity of the disease and the number of reactive T cells was found in KTRs. In 5 KTRs, also evaluated 10 mo after COVID-19, weak or absent antibody response was observed, whereas specific memory T-cell response was detected in all cases. Conclusion T-cell response persisted up to 3 mo post-symptom onset, even in KTRs in whom full immunosuppressive regimen was reinstated at recovery, and seems to be present up to 10 mo after infection. Our findings have implications in the understanding of the natural course of the disease in transplant patients and DPs.
Collapse
Affiliation(s)
- Dominique Bertrand
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Mouad Hamzaoui
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Laurent Drouot
- Normandy University, UNIROUEN, INSERM U1234, Rouen, France
| | - Julie Lamulle
- Normandy University, UNIROUEN, INSERM U1234, Rouen, France
| | - Mélanie Hanoy
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Stéphane Edet
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France.,ANIDER, Rouen, France
| | - Charlotte Laurent
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Ludivine Lebourg
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Isabelle Etienne
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Mathilde Lemoine
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Frank Le Roy
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Dorian Nezam
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Eleusis Mauger
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Olivier Boyer
- Normandy University, UNIROUEN, INSERM U1234, Rouen, France.,Department of Immunology and Biotherapies, Rouen University Hospital, Rouen, France
| | - Dominique Guerrot
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Sophie Candon
- Normandy University, UNIROUEN, INSERM U1234, Rouen, France.,Department of Immunology and Biotherapies, Rouen University Hospital, Rouen, France
| |
Collapse
|
26
|
Robust and Persistent B- and T-Cell Responses after COVID-19 in Immunocompetent and Solid Organ Transplant Recipient Patients. Viruses 2021; 13:v13112261. [PMID: 34835067 PMCID: PMC8621286 DOI: 10.3390/v13112261] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/05/2021] [Accepted: 11/07/2021] [Indexed: 12/18/2022] Open
Abstract
The development and persistence of SARS-CoV-2-specific immune response in immunocompetent (IC) and immunocompromised patients is crucial for long-term protection. Immune response to SARS-CoV-2 infection was analysed in 57 IC and 15 solid organ transplanted (TX) patients. Antibody responses were determined by ELISA and neutralization assay. T-cell response was determined by stimulation with peptide pools of the Spike, Envelope, Membrane, and Nucleocapsid proteins with a 20-h Activation Induced Marker (AIM) and 7-day lymphoproliferative assays. Antibody response was detected at similar levels in IC and TX patients. Anti-Spike IgG, IgA and neutralizing antibodies persisted for at least one year, while anti-Nucleocapsid IgG declined earlier. Patients with pneumonia developed higher antibody levels than patients with mild symptoms. Similarly, both rapid and proliferative T-cell responses were detected within the first two months after infection at comparable levels in IC and TX patients, and were higher in patients with pneumonia. T-cell response persisted for at least one year in both IC and TX patients. Spike, Membrane, and Nucleocapsid proteins elicited the major CD4+ and CD8+ T-cell responses, whereas the T-cell response to Envelope protein was negligible. After SARS-CoV-2 infection, antibody and T-cell responses develop rapidly and persist over time in both immunocompetent and transplanted patients.
Collapse
|
27
|
Kamińska D, Augustyniak-Bartosik H, Kościelska-Kasprzak K, Żabińska M, Bartoszek D, Poznański P, Kuriata-Kordek M, Kusztal M, Mazanowska O, Krajewska M. Comparing Humoral and Cellular Adaptive Immunity during Convalescent Phase of COVID-19 in Hemodialysis Patients and Kidney Transplant Recipients. J Clin Med 2021; 10:jcm10214833. [PMID: 34768356 PMCID: PMC8585082 DOI: 10.3390/jcm10214833] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/12/2021] [Accepted: 10/19/2021] [Indexed: 12/13/2022] Open
Abstract
Background. It is still unclear whether COVID-19 convalescent kidney transplant recipients (KTR) and hemodialysis (HD) patients can develop anti-SARS-CoV-2 adaptive immunity. The aim was to characterize and compare the immune response to the virus in HD patients and KTR. Methods. The study included 26 HD patients and 54 KTR—both convalescent (14 HD, 25 KTR) and unexposed. The immune response was assessed by determining the anti-SARS-CoV-2 antibodies in serum and specific T cell response via the interferon-gamma release assay (IGRA). Moreover, blood-morphology-derived parameters, immune cell phenotypes, and acute phase reactants were evaluated. Results. KRT and HD convalescents presented similar serum levels of anti-SARS-CoV-2 IgG and IgA. A negative correlation occurred between IgG and time after the infection was observed. There was a strong relationship between the prevalence of anti-SARS-CoV-2 cellular and humoral responses in both groups. Convalescent IGRA response was significantly higher in HD patients compared to KTR. Conclusions. HD patients and KTR develop humoral and cellular responses after COVID-19. The antibodies levels are similar in both groups of patients. SARS-CoV-2-reactive T cell response is stronger in HD patients compared to KTR. The SARS-CoV-2-specific IgG level decreases with time while IgA and a cellular response are maintained. IGRA proved to be a valuable test for the assessment of specific cellular immunity in immunocompromised HD patients and KTR.
Collapse
|
28
|
Lindemann M, Klisanin V, Thümmler L, Fisenkci N, Tsachakis-Mück N, Ditschkowski M, Schwarzkopf S, Klump H, Reinhardt HC, Horn PA, Koldehoff M. Humoral and Cellular Vaccination Responses against SARS-CoV-2 in Hematopoietic Stem Cell Transplant Recipients. Vaccines (Basel) 2021; 9:1075. [PMID: 34696183 PMCID: PMC8537291 DOI: 10.3390/vaccines9101075] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 01/14/2023] Open
Abstract
The cellular response to SARS-CoV-2 vaccination and infection in allogeneic hematopoietic stem cell transplant (HSCT) recipients is not yet clear. In the current study, HSCT recipients prior to and post vaccination were tested for SARS-CoV-2-specific humoral and cellular immunity. Antibodies against spike (S) 1 were assessed by Anti-SARS-CoV-2 IgG ELISA (Euroimmun). Cellular immunity was analyzed by an in house interferon-gamma ELISpot and T-SPOT.COVID (Oxford Immunotec), using altogether seven SARS-CoV-2-specific antigens. In 117 HSCT patients vaccinated twice, SARS-CoV-2 IgG antibodies were significantly higher than in HSCT controls pre vaccination (p < 0.0001). After the second vaccination, we observed a median antibody ratio of 4.7 and 68% positive results, whereas 35 healthy controls reached a median ratio of 9.0 and 100% positivity. ELISpot responses in patients were significantly (p < 0.001) reduced to ≤33% of the controls. After the second vaccination, female HSCT patients and female healthy controls showed significantly higher antibody responses than males (6.0 vs. 2.1 and 9.2 vs. 8.2, respectively; p < 0.05). Cellular immunity was diminished in patients irrespective of sex. In conclusion, especially male HSCT recipients showed impaired antibody responses after SARS-CoV-2 vaccination. Changing the vaccine schedule or composition could help increase vaccine responses.
Collapse
Affiliation(s)
- Monika Lindemann
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (L.T.); (N.F.); (S.S.); (H.K.); (P.A.H.)
| | - Vesna Klisanin
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (V.K.); (N.T.-M.); (M.D.); (H.C.R.); (M.K.)
| | - Laura Thümmler
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (L.T.); (N.F.); (S.S.); (H.K.); (P.A.H.)
| | - Neslinur Fisenkci
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (L.T.); (N.F.); (S.S.); (H.K.); (P.A.H.)
| | - Nikolaos Tsachakis-Mück
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (V.K.); (N.T.-M.); (M.D.); (H.C.R.); (M.K.)
| | - Markus Ditschkowski
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (V.K.); (N.T.-M.); (M.D.); (H.C.R.); (M.K.)
| | - Sina Schwarzkopf
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (L.T.); (N.F.); (S.S.); (H.K.); (P.A.H.)
| | - Hannes Klump
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (L.T.); (N.F.); (S.S.); (H.K.); (P.A.H.)
| | - Hans Christian Reinhardt
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (V.K.); (N.T.-M.); (M.D.); (H.C.R.); (M.K.)
| | - Peter A. Horn
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (L.T.); (N.F.); (S.S.); (H.K.); (P.A.H.)
| | - Michael Koldehoff
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (V.K.); (N.T.-M.); (M.D.); (H.C.R.); (M.K.)
| |
Collapse
|
29
|
Dęborska-Materkowska D, Kamińska D. The Immunology of SARS-CoV-2 Infection and Vaccines in Solid Organ Transplant Recipients. Viruses 2021; 13:1879. [PMID: 34578460 PMCID: PMC8473113 DOI: 10.3390/v13091879] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 12/12/2022] Open
Abstract
Since its outbreak in December 2019, the coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), led to an enormous rise in scientific response with an excess of COVID-19-related studies on the pathogenesis and potential therapeutic approaches. Solid organ transplant (SOT) recipients are a heterogeneous population with long-lasting immunosuppression as a joining element. Immunocompromised patients are a vulnerable population with a high risk of severe infections and an increased infection-related mortality rate. It was postulated that the hyperinflammatory state due to cytokine release syndrome during severe COVID-19 could be alleviated by immunosuppressive therapy in SOT patients. On the other hand, it was previously established that T cell-mediated immunity, which is significantly weakened in SOT recipients, is the main component of antiviral immune responses. In this paper, we present the current state of science on COVID-19 immunology in relation to solid organ transplantation with prospective therapeutic and vaccination strategies in this population.
Collapse
Affiliation(s)
- Dominika Dęborska-Materkowska
- Department of Transplantation Medicine, Nephrology and Internal Diseases, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland;
| | - Dorota Kamińska
- Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| |
Collapse
|
30
|
Reuken PA, Andreas N, Grunert PC, Glöckner S, Kamradt T, Stallmach A. T Cell Response After SARS-CoV-2 Vaccination in Immunocompromised Patients with Inflammatory Bowel Disease. J Crohns Colitis 2021; 16:251-258. [PMID: 34379729 PMCID: PMC8385945 DOI: 10.1093/ecco-jcc/jjab147] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Vaccination is a promising strategy to protect vulnerable groups like immunocompromised inflammatory bowel disease [IBD] patients from an infection with SARS-CoV-2. These patients may have lower immune responses. Little is known about the cellular and humoral immune response after a SARS-CoV-2 vaccination in IBD patients. METHODS Totals of 28 patients with IBD and 27 age- and sex-matched healthy controls were recruited at Jena University Hospital. Blood samples were taken before, after the first, and in a subgroup of 11 patients after second dose of a SARS-CoV-2 vaccination. Cellular immune response, including IFN-γ and TNF-α response and antibody titres, were analysed. RESULTS Overall, 71.4% of the IBD patients and 85.2% of the controls showed levels of anti-SARS-CoV-2 antibodies above the cutoff of 33.8 BAU/ml [p = 0.329] after the first dose. Even in the absence of SARS-CoV-2 antibodies, IBD patients showed significant T cell responses after first SARS-CoV-2 vaccination compared with healthy controls, which was not influenced by different immunosuppressive regimens. Associated with the vaccination, we could also detect a slight increase of the TNF production among SARS-CoV-2-reactive TH cells in healthy donorsn [HD] and IBD patients. After the second dose of vaccination, in IBD patients a further increase of humoral immune response in all but one patient was observed. CONCLUSIONS Already after the first dose of a SARS-CoV-2 vaccination, cellular immune response in IBD patients is comparable to controls, indicating a similar efficacy. However, close monitoring of long-term immunity in these patients should be considered.
Collapse
Affiliation(s)
- P A Reuken
- Jena University Hospital, Department of Internal Medicine IV, Jena, Germany,Address for Correspondence: Philipp Reuken, Department of Internal Medicine IV, Jena University Hospital, Friedrich Schiller University of Jena, Am Klinikum 1, 07747 Jena, Germany. Fax: +49-3641-9324222,
| | - N Andreas
- Institute for Immunology, Jena University Hospital, Jena, Germany
| | - P C Grunert
- Jena University Hospital, Department of Internal Medicine IV, Jena, Germany
| | - S Glöckner
- Institute for Medical Microbiology, Jena University Hospital, Jena, Germany
| | - T Kamradt
- Institute for Immunology, Jena University Hospital, Jena, Germany,Core Facility Cytometry, Jena University Hospital, Jena, Germany
| | - A Stallmach
- Jena University Hospital, Department of Internal Medicine IV, Jena, Germany
| |
Collapse
|
31
|
Fernández-Ruiz M, Olea B, Almendro-Vázquez P, Giménez E, Marcacuzco A, San Juan R, Justo I, Calvo-Pulido J, García-Sesma Á, Manrique A, Caso O, Cambra F, Talayero P, López-Medrano F, Remigia MJ, Ruiz-Merlo T, Parra P, Paz-Artal E, Jiménez C, Loinaz C, Navarro D, Laguna-Goya R, Aguado JM. T cell-mediated response to SARS-CoV-2 in liver transplant recipients with prior COVID-19. Am J Transplant 2021; 21:2785-2794. [PMID: 34092033 PMCID: PMC8222887 DOI: 10.1111/ajt.16708] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/11/2021] [Accepted: 05/30/2021] [Indexed: 01/25/2023]
Abstract
Whether immunosuppression impairs severe acute respiratory syndrome coronavirus 2-specific T cell-mediated immunity (SARS-CoV-2-CMI) after liver transplantation (LT) remains unknown. We included 31 LT recipients in whom SARS-CoV-2-CMI was assessed by intracellular cytokine staining (ICS) and interferon (IFN)-γ FluoroSpot assay after a median of 103 days from COVID-19 diagnosis. Serum SARS-CoV-2 IgG antibodies were measured by ELISA. A control group of nontransplant immunocompetent patients were matched (1:1 ratio) by age and time from diagnosis. Post-transplant SARS-CoV-2-CMI was detected by ICS in 90.3% (28/31) of recipients, with higher proportions for IFN-γ-producing CD4+ than CD8+ responses (93.5% versus 83.9%). Positive spike-specific and nucleoprotein-specific responses were found by FluoroSpot in 86.7% (26/30) of recipients each, whereas membrane protein-specific response was present in 83.3% (25/30). An inverse correlation was observed between the number of spike-specific IFN-γ-producing SFUs and time from diagnosis (Spearman's rho: -0.418; p value = .024). Two recipients (6.5%) failed to mount either T cell-mediated or IgG responses. There were no significant differences between LT recipients and nontransplant patients in the magnitude of responses by FluoroSpot to any of the antigens. Most LT recipients mount detectable-but declining over time-SARS-CoV-2-CMI after a median of 3 months from COVID-19, with no meaningful differences with immunocompetent patients.
Collapse
Affiliation(s)
- Mario Fernández-Ruiz
- Unit of Infectious Diseases, Hospital Universitario “12 de Octubre”, Instituto de Investigación Sanitaria Hospital “12 de Octubre” (imas12), Madrid, Spain
| | - Beatriz Olea
- Department of Microbiology, Hospital Clínico Universitario, Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
| | - Patricia Almendro-Vázquez
- Department of Immunology, Hospital Universitario “12 de Octubre”, Instituto de Investigación Sanitaria Hospital “12 de Octubre” (imas12), Madrid, Spain
| | - Estela Giménez
- Department of Microbiology, Hospital Clínico Universitario, Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
| | - Alberto Marcacuzco
- Department of General Surgery, Digestive Tract and Abdominal Organ Transplantation, Hospital Universitario “12 de Octubre”, Instituto de Investigación Sanitaria Hospital “12 de Octubre” (imas12), Madrid, Spain
| | - Rafael San Juan
- Unit of Infectious Diseases, Hospital Universitario “12 de Octubre”, Instituto de Investigación Sanitaria Hospital “12 de Octubre” (imas12), Madrid, Spain
- Department of Medicine, Universidad Complutense, Madrid, Spain
| | - Iago Justo
- Department of General Surgery, Digestive Tract and Abdominal Organ Transplantation, Hospital Universitario “12 de Octubre”, Instituto de Investigación Sanitaria Hospital “12 de Octubre” (imas12), Madrid, Spain
| | - Jorge Calvo-Pulido
- Department of General Surgery, Digestive Tract and Abdominal Organ Transplantation, Hospital Universitario “12 de Octubre”, Instituto de Investigación Sanitaria Hospital “12 de Octubre” (imas12), Madrid, Spain
| | - Álvaro García-Sesma
- Department of General Surgery, Digestive Tract and Abdominal Organ Transplantation, Hospital Universitario “12 de Octubre”, Instituto de Investigación Sanitaria Hospital “12 de Octubre” (imas12), Madrid, Spain
| | - Alejandro Manrique
- Department of General Surgery, Digestive Tract and Abdominal Organ Transplantation, Hospital Universitario “12 de Octubre”, Instituto de Investigación Sanitaria Hospital “12 de Octubre” (imas12), Madrid, Spain
| | - Oscar Caso
- Department of General Surgery, Digestive Tract and Abdominal Organ Transplantation, Hospital Universitario “12 de Octubre”, Instituto de Investigación Sanitaria Hospital “12 de Octubre” (imas12), Madrid, Spain
| | - Félix Cambra
- Department of General Surgery, Digestive Tract and Abdominal Organ Transplantation, Hospital Universitario “12 de Octubre”, Instituto de Investigación Sanitaria Hospital “12 de Octubre” (imas12), Madrid, Spain
| | - Paloma Talayero
- Department of Immunology, Hospital Universitario “12 de Octubre”, Instituto de Investigación Sanitaria Hospital “12 de Octubre” (imas12), Madrid, Spain
| | - Francisco López-Medrano
- Unit of Infectious Diseases, Hospital Universitario “12 de Octubre”, Instituto de Investigación Sanitaria Hospital “12 de Octubre” (imas12), Madrid, Spain
- Department of Medicine, Universidad Complutense, Madrid, Spain
| | - María José Remigia
- Department of Hematology, Hospital Clínico Universitario, Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
| | - Tamara Ruiz-Merlo
- Unit of Infectious Diseases, Hospital Universitario “12 de Octubre”, Instituto de Investigación Sanitaria Hospital “12 de Octubre” (imas12), Madrid, Spain
| | - Patricia Parra
- Unit of Infectious Diseases, Hospital Universitario “12 de Octubre”, Instituto de Investigación Sanitaria Hospital “12 de Octubre” (imas12), Madrid, Spain
| | - Estela Paz-Artal
- Department of Immunology, Hospital Universitario “12 de Octubre”, Instituto de Investigación Sanitaria Hospital “12 de Octubre” (imas12), Madrid, Spain
| | - Carlos Jiménez
- Department of General Surgery, Digestive Tract and Abdominal Organ Transplantation, Hospital Universitario “12 de Octubre”, Instituto de Investigación Sanitaria Hospital “12 de Octubre” (imas12), Madrid, Spain
- Department of Surgery, Universidad Complutense, Madrid, Spain
| | - Carmelo Loinaz
- Department of General Surgery, Digestive Tract and Abdominal Organ Transplantation, Hospital Universitario “12 de Octubre”, Instituto de Investigación Sanitaria Hospital “12 de Octubre” (imas12), Madrid, Spain
- Department of Surgery, Universidad Complutense, Madrid, Spain
| | - David Navarro
- Department of Microbiology, Hospital Clínico Universitario, Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
- School of Medicine, Universidad de Valencia, Valencia, Spain
| | - Rocío Laguna-Goya
- Department of Immunology, Hospital Universitario “12 de Octubre”, Instituto de Investigación Sanitaria Hospital “12 de Octubre” (imas12), Madrid, Spain
| | - José M. Aguado
- Unit of Infectious Diseases, Hospital Universitario “12 de Octubre”, Instituto de Investigación Sanitaria Hospital “12 de Octubre” (imas12), Madrid, Spain
- Department of Medicine, Universidad Complutense, Madrid, Spain
| |
Collapse
|
32
|
Kulkarni AV, Tevethia HV, Premkumar M, Arab JP, Candia R, Kumar K, Kumar P, Sharma M, Rao PN, Reddy DN. Impact of COVID-19 on liver transplant recipients-A systematic review and meta-analysis. EClinicalMedicine 2021; 38:101025. [PMID: 34278287 PMCID: PMC8276632 DOI: 10.1016/j.eclinm.2021.101025] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/21/2021] [Accepted: 06/28/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Immunosuppression and comorbidities increase the risk of severe coronavirus disease-2019 (COVID-19) in solid organ transplant (SOT) recipients. The outcomes of COVID-19 in liver transplant (LT) recipients remain unclear. We aimed to analyse the outcomes of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in LT recipients. METHODS The electronic databases were searched for articles published from 1 December 2019 to 20 May 2021 with MeSH terms COVID-19, SARS-CoV-2, and liver transplantation. Studies reporting outcomes in more than 10 LT recipients were included for analysis. LT vs non-LT patients with COVID-19 infection were compared for all-cause mortality, which was the primary outcome studied. We also evaluated the relation between the timing of COVID-19 infection post-LT (< one year vs > one year) and mortality. FINDINGS Eighteen articles reporting 1,522 COVID-19 infected LT recipients were included for the systematic review. The mean age (standard deviation [SD]) was 60·38 (5·24) years, and 68·5% were men. The mean time (SD) to COVID-19 infection was 5·72 (1·75) years. Based on 17 studies (I2 = 7·34) among 1,481 LT recipients, the cumulative incidence of mortality was 17·4% (95% confidence interval [CI], 15·4-19·6). Mortality was comparable between LT (n = 610) and non-LT (n = 239,704) patients, based on four studies (odds ratio [OR], 0·8 [0·6-1·08]; P = 0·14). Additionally, there was no significant difference in mortality between those infected within one year vs after one year of LT (OR, 1·5 [0·63-3·56]; P = 0·35). The cumulative incidence of graft dysfunction was 2·3% (1·3-4·1). Nearly 23% (20·71-25) of the LT patients developed severe COVID-19 infection. Before infection, 71% and 49% of patients were on tacrolimus and mycophenolate mofetil, respectively. Immunosuppression was modified in 55·9% (38·1-72·2) patients after COVID-19 infection. INTERPRETATION LT and non-LT patients with COVID-19 have a similar risk of adverse outcomes.
Collapse
Affiliation(s)
- Anand V. Kulkarni
- Department of Hepatology, Asian Institute of Gastroenterology, Hyderabad, India
| | | | | | - Juan Pablo Arab
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Roberto Candia
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Karan Kumar
- Department of Hepatology, Pacific Institute of Medical Sciences, Udaipur, India
| | - Pramod Kumar
- Department of Hepatology, Asian Institute of Gastroenterology, Hyderabad, India
| | - Mithun Sharma
- Department of Hepatology, Asian Institute of Gastroenterology, Hyderabad, India
| | - Padaki Nagaraja Rao
- Department of Hepatology, Asian Institute of Gastroenterology, Hyderabad, India
| | | |
Collapse
|