1
|
Rana MM, Haydel B, Carrara G, Gleason C, Mauldin J, Srivastava K, Florman SS, Aberg J, van Kesteren M, Mischka J, Carreño JM, Singh G, Mendu DR, Wajnberg A, Cordon-Cardo C, Krammer F, Simon V. Humoral SARS-CoV-2 vaccine responses are durable in solid organ transplant recipients with and without HIV. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.05.07.25327192. [PMID: 40385393 PMCID: PMC12083590 DOI: 10.1101/2025.05.07.25327192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Background Solid organ transplant (SOT) recipients may have a suboptimal humoral immune response to the coronavirus disease 2019 (COVID-19) vaccine, prompting the need for additional doses of vaccine for immunocompromised patients. However, data regarding immune responses to vaccination specifically in SOT recipients with well controlled HIV are lacking. Methods We conducted a prospective observational cohort single-center study of SOT recipients with and without HIV-1 who had received two doses of mRNA COVID-19 vaccine and were planning to receive additional doses. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) binding and neutralizing antibody responses were measured at several time points after vaccination. Findings Of the 122 SOT recipients enrolled, 44 (36%) were people with HIV (PWH). Overall, 65% (50/77) of all SOT recipients were seropositive prior to a third vaccine dose. Seropositive SOT recipients with HIV had comparable anti-spike antibody responses at baseline and over time to those without HIV. In addition, HIV status did not impact neutralizing titers in our SOT cohort. Twenty-seven participants were seronegative at baseline; three (11%) were participants with HIV. In addition, 78% (21/27) of participants seroconverted over the duration of the study; of those who remained seronegative, none had HIV, but all were on an antimetabolites. Interpretation HIV status did not impact longitudinal spike-binding antibody titers or neutralizing titers in SOT recipients. Research in context Evidence before this study Solid organ transplant (SOT) recipients may mount poor humoral immune responses to COVID-19 vaccines, prompting the need for additional vaccine doses in this patient population. Additional risk factors for poor immune response in this population have been described and include for example, age or use of certain immunosuppressant therapies. However, humoral responses to COVID-19 vaccine in SOT recipients with HIV have not previously been described. Added value of this study We conducted a prospective observational single center study of solid organ transplant recipients with and without HIV and measured SARS-CoV-2 binding and neutralizing antibody responses longitudinally. Our study results demonstrate that HIV status did not appear to be an additional risk factor that affected the durability of spike-antibody titers or neutralizing titers in SOT recipients over time. Implications of all the available evidence Well-controlled HIV infection is not an additional risk factor in SOT recipients when assessing responses to COVID-19 vaccine. Future studies should continue to focus on other risk factors, such as type of immunosuppressant therapies and timing of vaccination in relationship to transplant.
Collapse
|
2
|
Hoffmann S, Schrezenmeier E, Desmarets M, Halleck F, Durrbach A, Peters L, Tremmel AT, Seidel A, Führer M, Bachmann F, Schrezenmeier J, Greiner J, Körper S, Hofmann H, Ludwig C, Vieweg C, Jahrsdörfer B, Budde K, Schmidt M, Münch J, Joher N, Daguindau E, Grüner B, Brunotte G, Vauchy C, Seifried E, Bradshaw D, Estcourt LJ, Roberts DJ, Toussirot E, Rijnders B, Tiberghien P, Schrezenmeier H. Early, very high-titre convalescent plasma therapy in clinically vulnerable individuals with mild COVID-19: an international, randomised, open-label trial. EBioMedicine 2025; 113:105613. [PMID: 40020259 PMCID: PMC11919330 DOI: 10.1016/j.ebiom.2025.105613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/05/2025] [Accepted: 02/08/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND COVID-19 convalescent plasma (CCP) is a treatment option for COVID-19. This study investigated the safety and efficacy of early, very high-titre CCP in immunocompromised individuals with mild COVID-19. METHODS This randomised, controlled, open-label trial assessed CCP in immunocompromised patients (n = 120) with mild COVID-19 in 10 clinical trial centres across Germany, France, and the Netherlands. Patients were randomised 1:1 to receive either standard of care (SoC) alone (SoC group) or SoC and 2 units of CCP. Most patients (89.7%) had received ≥3 SARS-CoV-2 vaccinations. The primary endpoint was hospitalisation for progressive COVID-19 symptoms or death by day 28 after randomisation, analysed on a modified intention-to-treat basis (117 patients). The safety analysis included the full analysis set. The trial is registered with EudraCT 2021-006621-22, and ClinicalTrials.gov, NCT05271929. FINDINGS Between April 11, 2022 and November 27, 2023, 120 patients were enrolled. Patients in the CCP group received a median of 559 ml CCP from convalescent, vaccinated donors with very high levels of SARS-CoV-2 antibodies (median 81,810 IU/ml) at a median 4 days after symptom onset. The primary outcome occurred in 5/58 patients (8.6%) in the SoC group and in 0/59 patients (0%) in the CCP group, difference -8.6% (95% confidence interval of difference -19% to -0.80%; p-value 0.027; Fisher's exact test). The course of SARS-CoV-2 antibodies in the patients demonstrated a passive transfer of antibodies by the CCP, in particular neutralising effects against new SARS-CoV-2 variants. Whole genome sequencing of SARS-CoV-2 in patients during follow-up showed significant intra-host viral evolution, but without differences between groups. CCP was well tolerated. INTERPRETATION Early administration of high-titre CCP can prevent hospitalisation or death in immunocompromised patients with mild COVID-19. FUNDING Support-e project (European Union's Horizon 2020 Programme), German Federal Ministry of Education and Research, ZonMw, the Netherlands Organisation for Health Research and Development.
Collapse
Affiliation(s)
- Simone Hoffmann
- Institute of Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Maxime Desmarets
- Université Marie et Louis Pasteur, EFS, Inserm, RIGHT (UMR 1098), Besançon, France; CHU Besançon, Inserm, Centre d'Investigation Clinique (CIC 1431), Besançon, France
| | - Fabian Halleck
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Antoine Durrbach
- Department of Nephrology, AP-HP Hôpital Henri Mondor, Créteil, Île-de-France, France; INSERM UMR1186, Universite Paris Saclay, France
| | - Lynn Peters
- Division of Infectious Diseases, Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | | | - Alina Seidel
- Institute of Molecular Virology, Ulm University Medical Centre, Ulm, Germany
| | - Marita Führer
- Institute of Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Friederike Bachmann
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jens Schrezenmeier
- Division of Haematology, Oncology, and Cancer Immunology, Medical Department, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jochen Greiner
- Department of Internal Medicine, Diakonie Hospital Stuttgart, Stuttgart, Germany
| | - Sixten Körper
- Institute of Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Henrike Hofmann
- Institute of Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Carolin Ludwig
- Institute of Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Christiane Vieweg
- Institute of Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Bernd Jahrsdörfer
- Institute of Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Schmidt
- Institute of Transfusion Medicine and Immunohematology, German Red Cross Blood Transfusion Service Baden-Württemberg - Hessen, Frankfurt, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Centre, Ulm, Germany
| | - Nizar Joher
- Department of Nephrology, AP-HP Hôpital Henri Mondor, Créteil, Île-de-France, France
| | - Etienne Daguindau
- Université Marie et Louis Pasteur, EFS, Inserm, RIGHT (UMR 1098), Besançon, France; Haematology Department, CHU Besançon, Besançon, France
| | - Beate Grüner
- Division of Infectious Diseases, Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Gaëlle Brunotte
- CHU Besançon, Inserm, Centre d'Investigation Clinique (CIC 1431), Besançon, France
| | - Charline Vauchy
- Université Marie et Louis Pasteur, EFS, Inserm, RIGHT (UMR 1098), Besançon, France; CHU Besançon, Inserm, Centre d'Investigation Clinique (CIC 1431), Besançon, France
| | - Erhard Seifried
- Institute of Transfusion Medicine and Immunohematology, German Red Cross Blood Transfusion Service Baden-Württemberg - Hessen, Frankfurt, Germany
| | - Daniel Bradshaw
- Virus Reference Department, UK Health Security Agency, London, UK
| | - Lise J Estcourt
- NHS Blood and Transplant, Oxford, Oxfordshire, UK; Radcliffe Department of Medicine, University of Oxford, Oxford, Oxfordshire, UK
| | - David J Roberts
- NHS Blood and Transplant, Oxford, Oxfordshire, UK; Radcliffe Department of Medicine, University of Oxford, Oxford, Oxfordshire, UK
| | - Eric Toussirot
- Université Marie et Louis Pasteur, EFS, Inserm, RIGHT (UMR 1098), Besançon, France; CHU Besançon, Inserm, Centre d'Investigation Clinique (CIC 1431), Besançon, France
| | - Bart Rijnders
- Department of Internal Medicine, Section of Infectious Diseases and Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Pierre Tiberghien
- Université Marie et Louis Pasteur, EFS, Inserm, RIGHT (UMR 1098), Besançon, France; Etablissement Francais du Sang, La Plaine Saint-Denis, Île-de-France, France
| | - Hubert Schrezenmeier
- Institute of Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany.
| |
Collapse
|
3
|
Seo E, Shin EC, Jung MK. SARS-CoV-2 vaccine-elicited immune responses in solid organ transplant recipients. CLINICAL TRANSPLANTATION AND RESEARCH 2024; 38:247-256. [PMID: 39743229 PMCID: PMC11732761 DOI: 10.4285/ctr.24.0062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025]
Abstract
Solid organ transplant recipients (SOTRs) are considered a high-risk group for coronavirus disease 2019 (COVID-19). The adaptive immune responses generated by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination include humoral and cellular immune responses. Most studies on the SARS-CoV-2 vaccine have focused primarily on humoral immunity, but cellular immunity is vital for effectively controlling progression to severe COVID-19. In SOTRs, the vaccine-induced adaptive immune response is significantly attenuated compared to the response in healthy individuals. Nevertheless, vaccinated SOTRs exhibit a reduced rate and severity of SARS-CoV-2 infection. This review aims to provide a concise overview of the current understanding of SARS-CoV-2 vaccine-induced immune responses in SOTRs.
Collapse
Affiliation(s)
- Euri Seo
- The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Korea
| | - Eui-Cheol Shin
- The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Min Kyung Jung
- The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Korea
| |
Collapse
|
4
|
Malahe SRK, den Hartog Y, Rietdijk WJR, van Baarle D, de Kuiper R, Reijerkerk D, Ras AM, Geers D, Diavatopoulos DA, Messchendorp AL, van der Molen RG, Imhof C, Frölke SC, Bemelman FJ, Gansevoort RT, Hilbrands LB, Sanders JSF, GeurtsvanKessel CH, Kho MML, de Vries RD, Reinders MEJ, Baan CC. Repeated COVID-19 Vaccination Drives Memory T- and B-cell Responses in Kidney Transplant Recipients: Results From a Multicenter Randomized Controlled Trial. Transplantation 2024; 108:2420-2433. [PMID: 38902860 PMCID: PMC11581438 DOI: 10.1097/tp.0000000000005119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/30/2024] [Accepted: 05/16/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Insight into cellular immune responses to COVID-19 vaccinations is crucial for optimizing booster programs in kidney transplant recipients (KTRs). METHODS In an immunologic substudy of a multicenter randomized controlled trial (NCT05030974) investigating different repeated vaccination strategies in KTR who showed poor serological responses after 2 or 3 doses of an messenger RNA (mRNA)-based vaccine, we compared SARS-CoV-2-specific interleukin-21 memory T-cell and B-cell responses by enzyme-linked immunosorbent spot (ELISpot) assays and serum IgG antibody levels. Patients were randomized to receive: a single dose of mRNA-1273 (100 μg, n = 25), a double dose of mRNA-1273 (2 × 100 μg, n = 25), or a single dose of adenovirus type 26 encoding the SARS-CoV-2 spike glycoprotein (Ad26.COV2.S) (n = 25). In parallel, we also examined responses in 50 KTR receiving 100 μg mRNA-1273, randomized to continue (n = 25) or discontinue (n = 25) mycophenolate mofetil/mycophenolic acid. As a reference, the data were compared with KTR who received 2 primary mRNA-1273 vaccinations. RESULTS Repeated vaccination increased the seroconversion rate from 21% to 66% in all patients, which was strongly associated with enhanced levels of SARS-CoV-2-specific interleukin-21 memory T cells (odd ratio, 3.84 [1.89-7.78]; P < 0.001) and B cells (odd ratio, 35.93 [6.94-186.04]; P < 0.001). There were no significant differences observed in these responses among various vaccination strategies. In contrast to KTR vaccinated with 2 primary vaccinations, the number of antigen-specific memory B cells demonstrated potential for classifying seroconversion after repeated vaccination (area under the curve, 0.64; 95% confidence interval, 0.37-0.90; P = 0.26 and area under the curve, 0.95; confidence interval, 0.87-0.97; P < 0.0001, respectively). CONCLUSIONS Our study emphasizes the importance of virus-specific memory T- and B-cell responses for comprehensive understanding of COVID-19 vaccine efficacy among KTR.
Collapse
Affiliation(s)
- S. Reshwan K. Malahe
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Yvette den Hartog
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Wim J. R. Rietdijk
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Debbie van Baarle
- Department of Medical Microbiology and Infection Prevention, Virology and Immunology Research Group, University Medical Center Groningen, Groningen, the Netherlands
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Ronella de Kuiper
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Derek Reijerkerk
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Alicia M. Ras
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Daryl Geers
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Dimitri A. Diavatopoulos
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center Nijmegen, Nijmegen, the Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center Nijmegen, Nijmegen, the Netherlands
| | - A. Lianne Messchendorp
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Renate G. 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
| | - Céline Imhof
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Sophie C. Frölke
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Renal Transplant Unit, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Frederike J. Bemelman
- Renal Transplant Unit, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Ron T. Gansevoort
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Luuk B. Hilbrands
- Department of Nephrology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jan-Stephan F. Sanders
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Marcia M. L. Kho
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Rory D. de Vries
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marlies E. J. Reinders
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Carla C. Baan
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
5
|
Donadeu L, Gomez-Olles S, Casanova F, Torija A, Lopez-Meseguer M, Boada-Pérez M, Kervella D, Crespo E, Carrera-Muñoz C, Campos-Varela I, Castells L, Cortese MF, Esperalba J, Fernández-Naval C, Quintero J, Muñoz M, Agüero F, Gonzalez-Costello J, Lladó L, Favà A, Cañas L, del Mar de la Hoz-Caballero M, Meneghini M, Torres IB, Juvé M, Hafkamp FMJ, Vila M, Robles AG, Buzón MJ, Toapanta N, Zúñiga JM, Monforte V, Saez-Giménez B, Len O, Arcos IL, Miret E, Ariceta G, Pardo E, Martínez X, Moreso F, Bestard O. Role of SARS-CoV-2-specific memory B cells promoting immune protection after booster vaccination in solid organ transplantation. Front Immunol 2024; 15:1463769. [PMID: 39439787 PMCID: PMC11493670 DOI: 10.3389/fimmu.2024.1463769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/09/2024] [Indexed: 10/25/2024] Open
Abstract
Introduction Solid organ transplant (SOT) recipients display weak seroconversion and neutralizing antibody (NAb) responses after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination and remain at risk of severe coronavirus disease 2019 (COVID-19). While B-cell memory is the hallmark of serological immunity, its role in driving successful vaccine responses and providing immune protection in SOT patients remains unclear. Methods We investigated the function and interplay of SARS-CoV-2-specific memory B cells (mBc), different cytokineproducing T cells, and cross-reactive NAb in driving seroconversion and protection against COVID-19 in two cohorts. First, we studied a large cohort of 148 SOT recipients and 32 immunocompetent individuals who underwent several vaccinations. Subsequently, we assessed 25 SOT patients participating in a randomized controlled trial to compare two different immunosuppressive strategies for allowing successful seroconversion and memory-cell responses after booster vaccination. Results We corroborate previous findings that B- and T-cell memory responses are weaker and more delayed in SOT patients than in immunocompetent (IC) individuals; however, within the SOT cohort, we found that these responses are relatively stronger and more robust in patients not receiving mycophenolate mofetil (MMF)-based therapies. Anti- spike IgG titers strongly correlated with RBD-specific IgG-producing mBc, with both displaying broad viral cross reactivity. Prebooster SARS-CoV-2-specific mBc and IL-2- producing T cells accurately predicted Nab seroconversion (AUC, 0.828) and protection against severe COVID-19. While switching unresponsive SOT patients from calcineurin inhibitors (CNI)/MMF to a low-exposure CNI/mTOR-i regimen favored wider SARS-CoV-2-specific immune responses after a fourth booster vaccination, preformed RBD-specific mBc predicted NAb seroconversion. Discussion Our study adds new insights into the pathobiology of immune memory and highlights the pivotal role of SARS-CoV-2-specific mBc in promoting immune protection inSOT patients.
Collapse
Affiliation(s)
- Laura Donadeu
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Susana Gomez-Olles
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Laboratory of Pneumology, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Franc Casanova
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alba Torija
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Manuel Lopez-Meseguer
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Laboratory of Pneumology, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Lung Transplant Unit, Pneumology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Meritxell Boada-Pérez
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Laboratory of Pneumology, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Delphine Kervella
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Elena Crespo
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Claudia Carrera-Muñoz
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Isabel Campos-Varela
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Liver Unit, Vall d'Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Lluís Castells
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Liver Unit, Vall d'Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maria F. Cortese
- Microbiology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Juliana Esperalba
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Microbiology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Candela Fernández-Naval
- Microbiology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jesús Quintero
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Pediatric Hepatology and Liver Transplant Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marina Muñoz
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Pediatric Nephrology, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Fernando Agüero
- Department of Preventive Medicine and Epidemiology, Bellvitge University Hospital, Barcelona, Spain
| | - José Gonzalez-Costello
- Advanced Heart Failure and Heart Transplant Unit, Department of Cardiology, Hospital Universitari de Bellvitge, BIOHEART-Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute (IDIBELL), Universitat de Barcelona, Ciber Cardiovascular (CIBERCV), Barcelona, Spain
| | - Laura Lladó
- Liver Transplant Unit, Bellvitge University Hospital, Barcelona, Spain
| | - Alexandre Favà
- Kidney Transplant Unit, Bellvitge University Hospital, Barcelona, Spain
| | - Laura Cañas
- Kidney Transplant Unit, Nephrology department, Germans Trias i Pujol Hospital, Badalona, Spain
| | - María del Mar de la Hoz-Caballero
- Equipo de Atención Primaria Sant Rafael, Servei d'Atenció Primària (SAP) Muntanya, Gerència Territorial de Barcelona Ciutat, Institut Català de la Salut, Barcelona, Spain
| | - Maria Meneghini
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Irina B. Torres
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mariona Juvé
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - FMJ Hafkamp
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marta Vila
- Microbiology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alba G. Robles
- Infectious Diseases Department, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maria José Buzón
- Infectious Diseases Department, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Nestor Toapanta
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - José Miguel Zúñiga
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Víctor Monforte
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Laboratory of Pneumology, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Lung Transplant Unit, Pneumology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Berta Saez-Giménez
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Laboratory of Pneumology, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Lung Transplant Unit, Pneumology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Oscar Len
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Infectious Diseases, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ibai Los Arcos
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Infectious Diseases, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Enric Miret
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Urology Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Gema Ariceta
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Pediatric Nephrology, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Emma Pardo
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Xavier Martínez
- Department of Preventive Medicine and Epidemiology, Vall d’Hebron Hospital Universitari, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Francesc Moreso
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Oriol Bestard
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
6
|
Hauser D, Urda L, Lang C, Mittelholzer C, Otte F, Kipfer E, Zhang Y, Lett M, Schebitz C, Müller RU, Klimkait W, Klimkait T. Benefits of Repeated SARS-CoV-2 Vaccination and Virus-induced Cross-neutralization Potential in Immunocompromised Transplant Patients and Healthy Individuals. Open Forum Infect Dis 2024; 11:ofae527. [PMID: 39371367 PMCID: PMC11450466 DOI: 10.1093/ofid/ofae527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 09/06/2024] [Indexed: 10/08/2024] Open
Abstract
Background Current COVID-19 vaccines primarily target the Spike protein of defined virus variants, offering limited protection against emerging variants in immunocompetent individuals. Similarly, protective immunity following natural SARS-CoV-2 infection is variable and of short duration, raising concerns about immunocompromised individuals' vaccination strategies. Methods This prospective multicenter study examined 66 sera from 59 immunocompromised and 451 sera from 215 immunocompetent individuals from different pandemic periods. We establish and validate a live virus-based neutralization assay to determine the virus-inactivating potential against ancestral and current SARS-CoV-2 isolates. Results Our virus-based neutralization assay demonstrated superior performance over surrogate neutralization assays. We found strong but transient immunity after complete vaccination schemes, with single doses providing minimum neutralization, regardless of vaccine type. Combining vaccination-induced immunity with SARS-CoV-2 infection before or after vaccination yielded higher neutralizing titers than vaccination or infection alone, consistent across both study groups. Additional doses after a full vaccination course restored neutralization levels. Conclusions Potentially protective SARS-CoV-2 neutralization is reliably induced in immunocompromised individuals by prior attenuation of immunosuppression. First-generation vaccines protect against various SARS-CoV-2 variants in immunocompetent individuals, with effective cross-neutralization demonstrated up to the Delta variant but largely absent for later Omicron variants. Continuous vaccine updates are necessary to address emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- David Hauser
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Lorena Urda
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Christopher Lang
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Fabian Otte
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Enja Kipfer
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Yuepeng Zhang
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Martin Lett
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Roman-Ulrich Müller
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Wilfried Klimkait
- KfH-Nierenzentrum, Heilig-Geist-Gesundheitszentrum, Köln-Longerich, Germany
| | - Thomas Klimkait
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
7
|
Kehara H, Johnson-Whiting A, Yanagida R, Krishan K, Zhao H, Mishkin A, Cordova F, Criner GJ, Toyoda Y, Shigemura N. A Single-center Experience With >200 Lung Transplant Recipients With COVID-19 Infection. Transplant Direct 2024; 10:e1676. [PMID: 39220217 PMCID: PMC11365680 DOI: 10.1097/txd.0000000000001676] [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: 03/19/2024] [Revised: 04/22/2024] [Accepted: 05/15/2024] [Indexed: 09/04/2024] Open
Abstract
Background Although COVID-19 is no longer a declared global health emergency, data remain limited on the impact of COVID-19 in lung transplant recipients. Methods We identified lung transplant recipients who were diagnosed with COVID-19 from March 2020 through August 2022 in our institutional database and investigated clinical outcomes. We then analyzed outcomes based on date of COVID-19 diagnosis (first wave March 2020-October 2020; second wave November 2020-2021; third wave December 2021-September 2022) and compared these results. Results Of the 210 lung transplant recipients (median age 67; 67% men) enrolled, 140 (67%) required hospital admission. Among admitted recipients, 35 (25%) were intubated and 7 (5%) were placed on extracorporeal membrane oxygenation. Overall survival was 67.1% at 1 y and 59.0% at 2 y post-COVID-19 diagnosis. COVID-19 led to mortality in all 5 patients diagnosed during their index admission for lung transplantation. Although overall survival was significantly better in recipients with COVID-19 during the third wave, in-hospital mortality remained high (first wave 28%, second wave 38%, and 28% third wave). Vaccination (partially vaccinated versus none and fully vaccinated versus none) was the only significant protective factor for hospital admission, and age 70 y and older and partially vaccinated (versus none or fully vaccinated) were independent risk factors for in-hospital mortality. Conclusions Overall survival after COVID-19 infection in lung transplant recipients continues to improve; however, in-hospital mortality remains remarkably high. Vaccination appears to have been impactful in preventing hospital admission, but its impact on in-hospital mortality is still unclear. Further research is needed to better identify lung transplant recipients at high risk for mortality from COVID-19.
Collapse
Affiliation(s)
- Hiromu Kehara
- Division of Cardiovascular Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Ashley Johnson-Whiting
- Division of Cardiovascular Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Roh Yanagida
- Division of Cardiovascular Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Kewal Krishan
- Division of Cardiovascular Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Huaqing Zhao
- Department of Biomedical Education and Data Science, Center for Biostatistics and Epidemiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Aaron Mishkin
- Section of Infectious Diseases, Department of Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Francis Cordova
- Department of Thoracic Medicine and Surgery, Temple University Hospital, Philadelphia, PA
| | - Gerard J. Criner
- Department of Thoracic Medicine and Surgery, Temple University Hospital, Philadelphia, PA
| | - Yoshiya Toyoda
- Division of Cardiovascular Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Norihisa Shigemura
- Division of Cardiovascular Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
- Department of Thoracic Medicine and Surgery, Temple University Hospital, Philadelphia, PA
| |
Collapse
|
8
|
Wu L, Yang L, Qian X, Hu W, Wang S, Yan J. Mannan-Decorated Lipid Calcium Phosphate Nanoparticle Vaccine Increased the Antitumor Immune Response by Modulating the Tumor Microenvironment. J Funct Biomater 2024; 15:229. [PMID: 39194667 DOI: 10.3390/jfb15080229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024] Open
Abstract
With the rapid development of tumor immunotherapy, nanoparticle vaccines have attracted much attention as potential therapeutic strategies. A systematic review and analysis must be carried out to investigate the effect of mannose modification on the immune response to nanoparticles in regulating the tumor microenvironment, as well as to explore its potential clinical application in tumor therapy. Despite the potential advantages of nanoparticle vaccines in immunotherapy, achieving an effective immune response in the tumor microenvironment remains a challenge. Tumor immune escape and the overexpression of immunosuppressive factors limit its clinical application. Therefore, our review explored how to intervene in the immunosuppressive mechanism in the tumor microenvironment through the use of mannan-decorated lipid calcium phosphate nanoparticle vaccines to improve the efficacy of immunotherapy in patients with tumors and to provide new ideas and strategies for the field of tumor therapy.
Collapse
Affiliation(s)
- Liusheng Wu
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 19077, Singapore
| | - Lei Yang
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xinye Qian
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Wang Hu
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Shuang Wang
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Jun Yan
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
| |
Collapse
|
9
|
SeyedAlinaghi S, Dashti M, Afzalian A, Siami H, Ghasemzadeh A, Varshochi S, Parikhani SN, Amrollah MF, Nourian A, Mehraeen E, Dadras O. The immunologic outcomes and adverse events of COVID-19 vaccine booster dose in immunosuppressed people: A systematic review. Prev Med Rep 2024; 44:102778. [PMID: 38979481 PMCID: PMC11228787 DOI: 10.1016/j.pmedr.2024.102778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 05/20/2024] [Accepted: 05/28/2024] [Indexed: 07/10/2024] Open
Abstract
Introduction This study examines the efficacy and safety of three COVID-19 booster vaccines including mRNA-based vaccines (BNT162b2 (BioNTech/Pfizer) and/or mRNA-1273 (Moderna)), Non-Replicating Viral-Vector vaccines (ChAdOx1 nCoV-19 vaccine (AstraZeneca) and/or Ad26. COV2.S (Johnson & Johnson)), and Protein Subunit vaccine (SpikoGen) in immunosuppressed patients. Methods Relevant articles were systematically searched using medical subject heading (MeSH) and keywords "COVID-19" and "booster dose" or "booster vaccine" or ''fourth dose" in the online databases of PubMed, Embase, Scopus, and Web of Science. To identify eligible studies, a two-phase screening process was implemented. Initially, three researchers evaluated the studies based on the relevancy of the title and abstract. Results A total of 58 studies met the inclusion criteria and were included in this review. The findings suggest that booster doses offer greater protection against the disease than the primary dose. The study also compared various vaccine types, revealing that viral vector and nucleic acid vaccines outperformed inactivated vaccines. Results indicated that individuals receiving booster doses experienced superior outcomes compared to those without boosters. Vaccination against COVID-19 emerged as the most effective preventive measure against infection and symptom severity. Elevated antibody levels post-booster dose vaccination in the population signaled robust immune responses, underscoring the benefits of supplementary vaccine doses. Conclusion This systematic review highlights preliminary evidence supporting the immunologic outcomes and safety of COVID-19 vaccine boosters in enhancing immune responses against SARS-CoV-2. However, further research is needed to determine optimal timing intervals between primary vaccination series and boosters while considering global equity issues and variant-specific considerations.
Collapse
Affiliation(s)
- SeyedAhmad SeyedAlinaghi
- Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Dashti
- Department of Radiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arian Afzalian
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Haleh Siami
- School of Medicine, Islamic Azad University, Tehran, Iran
| | - Afsaneh Ghasemzadeh
- Department of Radiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanaz Varshochi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Masoomeh Fathi Amrollah
- Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| | - Anahid Nourian
- Department of Pharmacotherapy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Esmaeil Mehraeen
- Department of Health Information Technology, Khalkhal University of Medical Sciences, Khalkhal, Iran
| | - Omid Dadras
- Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Tharmaraj D, Boo I, O'Hara J, Sun S, Polkinghorne KR, Dendle C, Turner SJ, van Zelm MC, Drummer HE, Khoury G, Mulley WR. Serological responses and clinical outcomes following a three-dose primary COVID-19 vaccine schedule in kidney transplant recipients and people on dialysis. Clin Transl Immunology 2024; 13:e1523. [PMID: 39055736 PMCID: PMC11272417 DOI: 10.1002/cti2.1523] [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: 04/16/2024] [Revised: 06/10/2024] [Accepted: 07/13/2024] [Indexed: 07/27/2024] Open
Abstract
Objectives Despite vaccination strategies, people with chronic kidney disease, particularly kidney transplant recipients (KTRs), remained at high risk of poor COVID-19 outcomes. We assessed serological responses to the three-dose COVID-19 vaccine schedule in KTRs and people on dialysis, as well as seroresponse predictors and the relationship between responses and breakthrough infection. Methods Plasma from 30 KTRs and 17 people receiving dialysis was tested for anti-Spike receptor binding domain (RBD) IgG and neutralising antibodies (NAb) to the ancestral and Omicron BA.2 variant after Doses 2 and 3 of vaccination. Results After three doses, KTRs achieved lower anti-Spike RBD IgG levels (P < 0.001) and NAb titres than people receiving dialysis (P = 0.002). Seropositive cross-reactive Omicron neutralisation levels were achieved in 11/27 (40.7%) KTRs and 11/14 (78.6%) dialysis recipients. ChAdOx1/viral-vector vaccine type, higher mycophenolate dose (> 1 g per day) and lower absolute B-cell counts predicted poor serological responses in KTRs. ChAdOx-1 vaccine type and higher monocyte counts were negative predictors in dialysis recipients. Among ancestral NAb seroresponders, higher NAb levels positively correlated with higher Omicron neutralisation (R = 0.9, P < 0.001). More KTRs contracted SARS-CoV-2 infection (14/30; 47%) than dialysis recipients (5/17; 29%) and had more severe disease. Those with breakthrough infections had significantly lower median interdose incremental change in anti-Spike RBD IgG and ancestral NAb titres. Conclusion Serological responses to COVID-19 vaccines in KTRs lag behind their dialysis counterparts. KTRs remained at high risk of breakthrough infection after their primary vaccination schedule underlining their need for booster doses, strict infection prevention measures and close surveillance.
Collapse
Affiliation(s)
- Dhakshayini Tharmaraj
- Department of NephrologyMonash HealthClaytonVICAustralia
- Department of Medicine, Centre for Inflammatory DiseasesMonash UniversityMelbourneVICAustralia
| | - Irene Boo
- Burnet InstituteMelbourneVICAustralia
| | - Jessie O'Hara
- Department of Microbiology, Monash Biomedicine Discovery InstituteMonash UniversityMelbourneVICAustralia
| | - Shir Sun
- Burnet InstituteMelbourneVICAustralia
- Department of Immunology, School of Translational MedicineMonash University and Alfred HealthMelbourneVICAustralia
| | - Kevan R Polkinghorne
- Department of NephrologyMonash HealthClaytonVICAustralia
- Department of Medicine, Centre for Inflammatory DiseasesMonash UniversityMelbourneVICAustralia
- Department of Epidemiology and Preventive MedicineMonash UniversityMelbourneVICAustralia
| | - Claire Dendle
- Department of Medicine, Centre for Inflammatory DiseasesMonash UniversityMelbourneVICAustralia
- Monash Infectious DiseasesMonash HealthClaytonVICAustralia
| | - Stephen J Turner
- Department of Microbiology, Monash Biomedicine Discovery InstituteMonash UniversityMelbourneVICAustralia
| | - Menno C van Zelm
- Department of Immunology, School of Translational MedicineMonash University and Alfred HealthMelbourneVICAustralia
- Department of Immunology, Erasmus MCUniversity Medical CenterRotterdamThe Netherlands
| | - Heidi E Drummer
- Burnet InstituteMelbourneVICAustralia
- Department of Microbiology, Monash Biomedicine Discovery InstituteMonash UniversityMelbourneVICAustralia
- Department of Microbiology and ImmunologyUniversity of MelbourneMelbourneVictoriaAustralia
| | - Gabriela Khoury
- Burnet InstituteMelbourneVICAustralia
- Department of Microbiology, Monash Biomedicine Discovery InstituteMonash UniversityMelbourneVICAustralia
| | - William R Mulley
- Department of NephrologyMonash HealthClaytonVICAustralia
- Department of Medicine, Centre for Inflammatory DiseasesMonash UniversityMelbourneVICAustralia
| |
Collapse
|
11
|
Singer J, Tunbridge MJ, Shi B, Perkins GB, Chai CS, Salehi T, Sim BZ, Kireta S, Johnston JK, Akerman A, Milogiannakis V, Aggarwal A, Turville S, Hissaria P, Ying T, Wu H, Grubor-Bauk B, Coates PT, Chadban SJ. Dietary Inulin to Improve SARS-CoV-2 Vaccine Response in Kidney Transplant Recipients: The RIVASTIM-Inulin Randomised Controlled Trial. Vaccines (Basel) 2024; 12:608. [PMID: 38932337 PMCID: PMC11209582 DOI: 10.3390/vaccines12060608] [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: 05/02/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Kidney transplant recipients are at an increased risk of hospitalisation and death from SARS-CoV-2 infection, and standard two-dose vaccination schedules are typically inadequate to generate protective immunity. Gut dysbiosis, which is common among kidney transplant recipients and known to effect systemic immunity, may be a contributing factor to a lack of vaccine immunogenicity in this at-risk cohort. The gut microbiota modulates vaccine responses, with the production of immunomodulatory short-chain fatty acids by bacteria such as Bifidobacterium associated with heightened vaccine responses in both observational and experimental studies. As SCFA-producing populations in the gut microbiota are enhanced by diets rich in non-digestible fibre, dietary supplementation with prebiotic fibre emerges as a potential adjuvant strategy to correct dysbiosis and improve vaccine-induced immunity. In a randomised, double-bind, placebo-controlled trial of 72 kidney transplant recipients, we found dietary supplementation with prebiotic inulin for 4 weeks before and after a third SARS-CoV2 mRNA vaccine to be feasible, tolerable, and safe. Inulin supplementation resulted in an increase in gut Bifidobacterium, as determined by 16S RNA sequencing, but did not increase in vitro neutralisation of live SARS-CoV-2 virus at 4 weeks following a third vaccination. Dietary fibre supplementation is a feasible strategy with the potential to enhance vaccine-induced immunity and warrants further investigation.
Collapse
Affiliation(s)
- Julian Singer
- Department of Renal Medicine, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia; (J.S.); (T.Y.); (H.W.)
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia;
| | - Matthew J. Tunbridge
- Central and Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (M.J.T.); (T.S.); (B.Z.S.); (S.K.); (J.K.J.); (P.T.C.)
| | - Bree Shi
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia;
| | - Griffith B. Perkins
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia; (G.B.P.); (C.S.C.); (P.H.); (B.G.-B.)
- Immunology Directorate, SA Pathology, Adelaide, SA 5000, Australia
| | - Cheng Sheng Chai
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia; (G.B.P.); (C.S.C.); (P.H.); (B.G.-B.)
| | - Tania Salehi
- Central and Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (M.J.T.); (T.S.); (B.Z.S.); (S.K.); (J.K.J.); (P.T.C.)
| | - Beatrice Z. Sim
- Central and Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (M.J.T.); (T.S.); (B.Z.S.); (S.K.); (J.K.J.); (P.T.C.)
| | - Svjetlana Kireta
- Central and Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (M.J.T.); (T.S.); (B.Z.S.); (S.K.); (J.K.J.); (P.T.C.)
| | - Julie K. Johnston
- Central and Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (M.J.T.); (T.S.); (B.Z.S.); (S.K.); (J.K.J.); (P.T.C.)
| | - Anouschka Akerman
- Kirby Institute, University of New South Wales, Sydney, NSW 2052, Australia; (A.A.); (V.M.); (A.A.); (S.T.)
| | - Vanessa Milogiannakis
- Kirby Institute, University of New South Wales, Sydney, NSW 2052, Australia; (A.A.); (V.M.); (A.A.); (S.T.)
| | - Anupriya Aggarwal
- Kirby Institute, University of New South Wales, Sydney, NSW 2052, Australia; (A.A.); (V.M.); (A.A.); (S.T.)
| | - Stuart Turville
- Kirby Institute, University of New South Wales, Sydney, NSW 2052, Australia; (A.A.); (V.M.); (A.A.); (S.T.)
| | - Pravin Hissaria
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia; (G.B.P.); (C.S.C.); (P.H.); (B.G.-B.)
- Department of Immunology and Allergy, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Tracey Ying
- Department of Renal Medicine, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia; (J.S.); (T.Y.); (H.W.)
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia;
| | - Huiling Wu
- Department of Renal Medicine, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia; (J.S.); (T.Y.); (H.W.)
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia;
| | - Branka Grubor-Bauk
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia; (G.B.P.); (C.S.C.); (P.H.); (B.G.-B.)
- Viral Immunology Group, Basil Hetzel Institute for Translational Health Research, University of Adelaide, Adelaide, SA 5011, Australia
| | - P. Toby Coates
- Central and Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (M.J.T.); (T.S.); (B.Z.S.); (S.K.); (J.K.J.); (P.T.C.)
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia; (G.B.P.); (C.S.C.); (P.H.); (B.G.-B.)
| | - Steven J. Chadban
- Department of Renal Medicine, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia; (J.S.); (T.Y.); (H.W.)
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia;
| |
Collapse
|
12
|
Kim AHJ. Additional SARS-CoV-2 vaccine doses: a little is better than none. THE LANCET. RHEUMATOLOGY 2024; 6:e330-e331. [PMID: 38734020 DOI: 10.1016/s2665-9913(24)00119-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024]
Affiliation(s)
- Alfred H J Kim
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
13
|
Kawashiro K, Suzuki R, Nogimori T, Tsujino S, Iwahara N, Hirose T, Okada K, Yamamoto T, Fukuhara T, Hotta K, Shinohara N. Neutralizing antibody responses and cellular responses against SARS-CoV-2 Omicron subvariants after mRNA SARS-CoV-2 vaccination in kidney transplant recipients. Sci Rep 2024; 14:12176. [PMID: 38806644 PMCID: PMC11133393 DOI: 10.1038/s41598-024-63147-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: 01/12/2024] [Accepted: 05/24/2024] [Indexed: 05/30/2024] Open
Abstract
Although the mRNA SARS-CoV-2 vaccine has improved the mortality rate in the general population, its efficacy against rapidly mutating virus strains, especially in kidney transplant recipients, remains unclear. We examined the anti-SARS-CoV-2 spike protein IgG antibody and neutralizing antibody titers and cellular immunity against B.1.1, BA.1, and BA.5 antigens in 73 uninfected kidney recipients and 16 uninfected healthy controls who received three doses of an mRNA SARS-CoV-2 vaccine. The IgG antibody titers were significantly lower in recipients than in healthy controls. Similarly, neutralizing antibody titers against three viral variants were significantly lower in recipients. When the virus was mutated, the neutralizing antibody titers decreased significantly in both groups. In cellular immunity analysis, the number of spike-specific CD8 + non-naïve T cells against three variants significantly decreased in recipients. Conversely, the frequency of spike-specific Th2 CD4 + T-cells in recipients was higher than that in healthy controls. Nineteen recipients and six healthy controls also received a bivalent omicron-containing booster vaccine, leading to increase IgG and neutralizing antibody titers in both groups. After that, eleven recipients and five healthy controls received XBB.1.5 monovalent vaccines, increasing the neutralizing antibody titers against not only XBB.1.5, but also EG.5.1 and BA.2.86 antigens in kidney recipients. Although kidney recipients did not gain sufficient immunity against Omicron BA.5 with the third dose of vaccine, humoral response against mutant SARS-CoV-2 lineages significantly increased after bivalent Omicron-containing booster vaccine and the XBB.1.5 monovalent vaccine. Therefore, it is important for kidney recipients to continue to administer updated vaccines.
Collapse
Affiliation(s)
- Keita Kawashiro
- Department of Urology, Hokkaido University Hospital, Sapporo, Japan
| | - Rigel Suzuki
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development: HU-IVReD, Hokkaido University, Sapporo, Japan
| | - Takuto Nogimori
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Japan
| | - Shuhei Tsujino
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Naoya Iwahara
- Department of Urology, Hokkaido University Hospital, Sapporo, Japan
| | - Takayuki Hirose
- Department of Urology, Hokkaido University Hospital, Sapporo, Japan
| | - Kazufumi Okada
- Data Science Center, Promotion Unit, Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Sapporo, Japan
| | - Takuya Yamamoto
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Japan.
- Laboratory of Aging and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan.
- Department of Virology and Immunology, Graduate School of Medicine, Osaka University, Suita, Japan.
| | - Takasuke Fukuhara
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan.
- Institute for Vaccine Research and Development: HU-IVReD, Hokkaido University, Sapporo, Japan.
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Suita, Japan.
- AMED-CREST, Japan Agency for Medical Research and Development (AMED), Tokyo, Japan.
| | - Kiyohiko Hotta
- Department of Urology, Hokkaido University Hospital, Sapporo, Japan.
| | - Nobuo Shinohara
- Department of Urology, Hokkaido University Hospital, Sapporo, Japan
| |
Collapse
|
14
|
New J, Cham J, Smith L, Puglisi L, Huynh T, Kurian S, Bagsic S, Fielding R, Hong L, Reddy P, Eum KS, Martin A, Barrick B, Marsh C, Quigley M, Nicholson LJ, Pandey AC. Effects of antineoplastic and immunomodulating agents on postvaccination SARS-CoV-2 breakthrough infections, antibody response, and serological cytokine profile. J Immunother Cancer 2024; 12:e008233. [PMID: 38296596 PMCID: PMC10831464 DOI: 10.1136/jitc-2023-008233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2023] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Despite immunization, patients on antineoplastic and immunomodulating agents have a heightened risk of COVID-19 infection. However, accurately attributing this risk to specific medications remains challenging. METHODS An observational cohort study from December 11, 2020 to September 22, 2022, within a large healthcare system in San Diego, California, USA was designed to identify medications associated with greatest risk of postimmunization SARS-CoV-2 infection. Adults prescribed WHO Anatomical Therapeutic Chemical (ATC) classified antineoplastic and immunomodulating medications were matched (by age, sex, race, and number of immunizations) with control patients not prescribed these medications yielding a population of 26 724 patients for analysis. From this population, 218 blood samples were collected from an enrolled subset to assess serological response and cytokine profile in relation to immunization. RESULTS Prescription of WHO ATC classified antineoplastic and immunomodulatory agents was associated with elevated postimmunization SARS-CoV-2 infection risk (HR 1.50, 95% CI 1.38 to 1.63). While multiple immunization doses demonstrated a decreased association with postimmunization SARS-CoV-2 infection risk, antineoplastic and immunomodulatory treated patients with four doses remained at heightened risk (HR 1.23, 95% CI 1.06 to 1.43). Risk variation was identified among medication subclasses, with PD-1/PD-L1 inhibiting monoclonal antibodies, calcineurin inhibitors, and CD20 monoclonal antibody inhibitors identified to associate with increased risk of postimmunization SARS-CoV-2 infection. Antineoplastic and immunomodulatory treated patients also displayed a reduced IgG antibody response to SARS-CoV-2 epitopes alongside a unique serum cytokine profile. CONCLUSIONS Antineoplastic and immunomodulating medications associate with an elevated risk of postimmunization SARS-CoV-2 infection in a drug-specific manner. This comprehensive, unbiased analysis of all WHO ATC classified antineoplastic and immunomodulating medications identifies medications associated with greatest risk. These findings are crucial in guiding and refining vaccination strategies for patients prescribed these treatments, ensuring optimized protection for this susceptible population in future COVID-19 variant surges and potentially for other RNA immunization targets.
Collapse
Affiliation(s)
- Jacob New
- Medicine, Scripps Health, La Jolla, California, USA
- Scripps Research Translational Institute, La Jolla, California, USA
| | - Jason Cham
- Scripps Research Translational Institute, La Jolla, California, USA
| | - Lana Smith
- Scripps Research Translational Institute, La Jolla, California, USA
| | - Leah Puglisi
- Medicine, Scripps Health, La Jolla, California, USA
| | - Tridu Huynh
- Scripps Research Translational Institute, La Jolla, California, USA
- Division of Hematology/Oncology, University of California, La Jolla, California, USA
| | - Sunil Kurian
- Scripps Organ Transplantation Research & Biorepository, Scripps Health, La Jolla, California, USA
| | | | - Russel Fielding
- Strategy & Planning, Scripps Health, La Jolla, California, USA
| | - Lee Hong
- Medicine, Scripps Health, La Jolla, California, USA
- Scripps Research Translational Institute, La Jolla, California, USA
| | - Priya Reddy
- Medicine, Scripps Health, La Jolla, California, USA
| | - Ki Suk Eum
- Medicine, Scripps Health, La Jolla, California, USA
- Rheumatology, Veterans Administration Pacific Islands Healthcare System, Honolulu, Hawaii, USA
| | - Allison Martin
- Scripps Organ Transplantation Research & Biorepository, Scripps Health, La Jolla, California, USA
| | - Bethany Barrick
- Scripps Organ Transplantation Research & Biorepository, Scripps Health, La Jolla, California, USA
| | - Christopher Marsh
- Scripps Organ Transplantation Research & Biorepository, Scripps Health, La Jolla, California, USA
| | | | - Laura J Nicholson
- Medicine, Scripps Health, La Jolla, California, USA
- Scripps Research Translational Institute, La Jolla, California, USA
| | - Amitabh C Pandey
- Scripps Research Translational Institute, La Jolla, California, USA
- Medicine, Section of Cardiology, Tulane University, New Orleans, Louisiana, USA
- Medicine, Southeast Veterans Health Care System, New Orleans, Louisiana, USA
| |
Collapse
|
15
|
Schrezenmeier E, Dörner T, Halleck F, Budde K. Cellular Immunobiology and Molecular Mechanisms in Alloimmunity-Pathways of Immunosuppression. Transplantation 2024; 108:148-160. [PMID: 37309030 DOI: 10.1097/tp.0000000000004646] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Current maintenance immunosuppression commonly comprises a synergistic combination of tacrolimus as calcineurin inhibitor (CNI), mycophenolic acid, and glucocorticoids. Therapy is often individualized by steroid withdrawal or addition of belatacept or inhibitors of the mechanistic target of rapamycin. This review provides a comprehensive overview of their mode of action, focusing on the cellular immune system. The main pharmacological action of CNIs is suppression of the interleukin-2 pathway that leads to inhibition of T cell activation. Mycophenolic acid inhibits the purine pathway and subsequently diminishes T and B cell proliferation but also exerts a variety of effects on almost all immune cells, including inhibition of plasma cell activity. Glucocorticoids exert complex regulation via genomic and nongenomic mechanisms, acting mainly by downregulating proinflammatory cytokine signatures and cell signaling. Belatacept is potent in inhibiting B/T cell interaction, preventing formation of antibodies; however, it lacks the potency of CNIs in preventing T cell-mediated rejections. Mechanistic target of rapamycin inhibitors have strong antiproliferative activity on all cell types interfering with multiple metabolic pathways, partly explaining poor tolerability, whereas their superior effector T cell function might explain their benefits in the case of viral infections. Over the past decades, clinical and experimental studies provided a good overview on the underlying mechanisms of immunosuppressants. However, more data are needed to delineate the interaction between innate and adaptive immunity to better achieve tolerance and control of rejection. A better and more comprehensive understanding of the mechanistic reasons for failure of immunosuppressants, including individual risk/benefit assessments, may permit improved patient stratification.
Collapse
Affiliation(s)
- Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Academy, Clinician Scientist Program Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas Dörner
- Department of Rheumatology and Clinical Immunology - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Deutsches Rheumaforschungszentrum (DRFZ), Berlin, Germany
| | - Fabian Halleck
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
16
|
Herrera S, Aguado JM, Candel FJ, Cordero E, Domínguez-Gil B, Fernández-Ruiz M, Los Arcos I, Len Ò, Marcos MÁ, Muñez E, Muñoz P, Rodríguez-Goncer I, Sánchez-Céspedes J, Valerio M, Bodro M. Executive summary of the consensus statement of the group for the study of infection in transplantation and other immunocompromised host (GESITRA-IC) of the Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC) on the treatment of SARS-CoV-2 infection in solid organ transplant recipients. Transplant Rev (Orlando) 2023; 37:100788. [PMID: 37591117 DOI: 10.1016/j.trre.2023.100788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 08/19/2023]
Affiliation(s)
- Sabina Herrera
- Department of Infectious Diseases, Hospital Clínic, IDIBAPS (Institut D'Investigacions Biomèdiques Agust Pi I Sunyer), Universitat de Barcelona, Barcelona, Spain
| | - Jose M Aguado
- Infectious Diseases Unit, Hospital Universitario 12 de Octubre (Madrid), Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Francisco Javier Candel
- Clinical Microbiology & Infectious Diseases, Transplant Coordination, Hospital Clínico Universitario San Carlos, Madrid 28040, Spain; Department of Clinical Microbiology and Infectious Diseases, Hospital Clínico San Carlos, Madrid, Spain
| | - Elisa Cordero
- Infectious Diseases Unit, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina Sevilla, Sevilla, Spain
| | | | - Mario Fernández-Ruiz
- Infectious Diseases Unit, Hospital Universitario 12 de Octubre (Madrid), Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Ibai Los Arcos
- Infectious Diseases Department, Hospital Universitari Vall D'Hebron, Barcelona, Spain
| | - Òscar Len
- Infectious Diseases Department, Hospital Universitari Vall D'Hebron, Barcelona, Spain
| | | | - Elena Muñez
- Infectious Diseases Unit, Internal Medicine Department, University Hospital Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Patricia Muñoz
- Department of Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, 9 Madrid, Spain
| | - Isabel Rodríguez-Goncer
- Infectious Diseases Unit, Hospital Universitario 12 de Octubre (Madrid), Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Javier Sánchez-Céspedes
- Infectious Diseases Unit, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina Sevilla, Sevilla, Spain
| | - Maricela Valerio
- Department of Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, 9 Madrid, Spain
| | - Marta Bodro
- Department of Infectious Diseases, Hospital Clínic, IDIBAPS (Institut D'Investigacions Biomèdiques Agust Pi I Sunyer), Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
17
|
Yu B, Tamargo C, Brennan DC, Kant S. Measures to Increase Immunogenicity of SARS-CoV-2 Vaccines in Solid Organ Transplant Recipients: A Narrative Review. Vaccines (Basel) 2023; 11:1755. [PMID: 38140160 PMCID: PMC10748337 DOI: 10.3390/vaccines11121755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/17/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
Purpose of review: To review the data on the immunogenicity of COVID-19 vaccines, administered by different strategies, in solid organ transplant recipients (SOTRs). Recent findings: COVID-19 booster vaccines were given to SOTRs as a widespread practice in many transplant centers, mostly as the third and/or fourth dose in an extended vaccine series, with a significantly improved humoral response compared with the initial two-dose scheme. However, one-third of SOTRs remained unresponsive, despite these boosters. Next steps: Vaccination with standard dosing remains the most feasible strategy for attaining protection against COVID-19. Additional booster doses and temporarily holding or reducing mycophenolate mofetil/mycophenolic acid may provide immunogenicity to vaccines, according to recent studies demonstrating some efficacy with these measures. Preexposure prophylaxis with monoclonal antibodies showed benefit in immunocompromised patients but is no longer recommended by the National Institutes of Health (NIH) due to diminished efficacy against Omicron and recent variants. Screening for the presence and titers of SARS-CoV-2-specific antibodies in SOTRs is not recommended in most clinical settings. T cell-based techniques are needed to evaluate vaccine efficacy and risk of infection. As SARS-CoV-2 continues to evolve, new vaccines based on conservative protein component/complexes of the COVID virus, in addition to its spike protein, are warranted to offer prolonged protection.
Collapse
Affiliation(s)
- Bo Yu
- Department of Medicine, University of Maryland Medical Center, Midtown Campus, Baltimore, MD 21201, USA;
| | - Christina Tamargo
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| | - Daniel C. Brennan
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
- Comprehensive Transplant Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sam Kant
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
- Comprehensive Transplant Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
18
|
Thole LML, Tóth L, Proß V, Siegle J, Stahl C, Hermsdorf G, Knabe A, Winkler A, Schrezenmeier E, Ludwig C, Eckert C, Eggert A, Schrezenmeier H, Sattler A, Schulte JH, Kotsch K. Impact of a booster dose on SARS-CoV2 mRNA vaccine-specific humoral-, B- and T cell immunity in pediatric stem cell transplant recipients. Front Immunol 2023; 14:1239519. [PMID: 37942315 PMCID: PMC10628529 DOI: 10.3389/fimmu.2023.1239519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 10/09/2023] [Indexed: 11/10/2023] Open
Abstract
Stem cell transplant recipients (SCTR) are imperiled to increased risks after SARS-CoV2 infection, supporting the need for effective vaccination strategies for this vulnerable group. With respect to pediatric patients, data on immunogenicity of SARS-CoV2 mRNA-based vaccination is limited. We therefore comprehensively examined specific humoral, B- and T cell responses in a cohort of 2-19 year old SCTR after the second and third vaccine dose. Only after booster vaccination, transplant recipients reached similar levels of vaccine-specific IgG, IgA and neutralizing antibodies against omicron variant as age-matched controls. Although frequencies of SARS-CoV2 specific B cells increased after the third dose, they were still fourfold reduced in patients compared to controls. Overall, the majority of individuals enrolled mounted SARS-CoV2 Spike protein-specific CD4+ T helper cell responses with patients showing significantly higher portions than controls after the third dose. With respect to functionality, however, SCTR were characterized by reduced frequencies of specific interferon gamma producing CD4+ T cells, along with an increase in IL-2 producers. In summary, our data identify distinct quantitative and qualitative impairments within the SARS-CoV2 vaccination specific B- and CD4+ T cell compartments. More importantly, humoral analyses highlight the need for a booster vaccination of SCTR particularly for development of neutralizing antibodies.
Collapse
Affiliation(s)
- Linda Marie Laura Thole
- Department of General and Visceral Surgery, Charite-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Laura Tóth
- Department of General and Visceral Surgery, Charite-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Vanessa Proß
- Department of General and Visceral Surgery, Charite-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Janine Siegle
- Department of General and Visceral Surgery, Charite-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Carolin Stahl
- Department of General and Visceral Surgery, Charite-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Georg Hermsdorf
- Department of Pediatric Oncology and Hematology, Charite-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Annette Knabe
- Department of Pediatric Oncology and Hematology, Charite-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Annika Winkler
- Department of Pediatric Oncology and Hematology, Charite-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charite-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin Institute of Health (BIH) Academy, Clinician Scientist Program Universitätsmedizin Berlin, Berlin, Germany
| | - Carolin Ludwig
- Institute of Transfusion Medicine, Ulm University, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg – Hessen and University Hospital Ulm, Ulm, Germany
| | - Cornelia Eckert
- Department of Pediatric Oncology and Hematology, Charite-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Angelika Eggert
- Department of Pediatric Oncology and Hematology, Charite-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Hubert Schrezenmeier
- Institute of Transfusion Medicine, Ulm University, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg – Hessen and University Hospital Ulm, Ulm, Germany
| | - Arne Sattler
- Department of General and Visceral Surgery, Charite-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Johannes H. Schulte
- Department of Pediatric Hematology and Oncology, University Children’s Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Katja Kotsch
- Department of General and Visceral Surgery, Charite-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
19
|
Mikhailov M, Budde K, Halleck F, Eleftheriadis G, Naik MG, Schrezenmeier E, Bachmann F, Choi M, Duettmann W, von Hoerschelmann E, Koch N, Liefeldt L, Lücht C, Straub-Hohenbleicher H, Waiser J, Weber U, Zukunft B, Osmanodja B. COVID-19 Outcomes in Kidney Transplant Recipients in a German Transplant Center. J Clin Med 2023; 12:6103. [PMID: 37763043 PMCID: PMC10531713 DOI: 10.3390/jcm12186103] [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: 08/10/2023] [Revised: 09/02/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Kidney transplant recipients (KTRs) show higher morbidity and mortality from COVID-19 than the general population and have an impaired response to vaccination. We analyzed COVID-19 incidence and clinical outcomes in a single-center cohort of approximately 2500 KTRs. Between 1 February 2020 and 1 July 2022, 578 KTRs were infected with SARS-CoV-2, with 25 (4%) recurrent infections. In total, 208 KTRs (36%) were hospitalized, and 39 (7%) died. Among vaccinated patients, infection with the Omicron variant had a mortality of 2%. Unvaccinated patients infected with the Omicron variant showed mortality (9% vs. 11%) and morbidity (hospitalization 52% vs. 54%, ICU admission 12% vs. 18%) comparable to the pre-Omicron era. Multivariable analysis revealed that being unvaccinated (OR = 2.15, 95% CI [1.38, 3.35]), infection in the pre-Omicron era (OR = 3.06, 95% CI [1.92, 4.87]), and higher patient age (OR = 1.04, 95% CI [1.03, 1.06]) are independent risk factors for COVID-19 hospitalization, whereas a steroid-free immunosuppressive regimen was found to reduce the risk of COVID-19 hospitalization (OR = 0.51, 95% CI [0.33, 0.79]). This suggests that both virological changes in the Omicron variant and vaccination reduce the risk for morbidity and mortality from COVID-19 in KTRs. Our data extend the knowledge from the general population to KTRs and provide important insights into outcomes during the Omicron era.
Collapse
Affiliation(s)
- Michael Mikhailov
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
- Clinic for Anaesthesiology and Intensive Care Medicine, Charité–Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Fabian Halleck
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Georgios Eleftheriadis
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Marcel G. Naik
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Friederike Bachmann
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Mira Choi
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Wiebke Duettmann
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Ellen von Hoerschelmann
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Nadine Koch
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Lutz Liefeldt
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Christian Lücht
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Henriette Straub-Hohenbleicher
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Johannes Waiser
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Ulrike Weber
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Bianca Zukunft
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| | - Bilgin Osmanodja
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (K.B.); (F.H.); (G.E.); (M.G.N.); (E.S.); (F.B.); (M.C.); (W.D.); (E.v.H.); (N.K.); (L.L.); (C.L.); (H.S.-H.); (J.W.); (U.W.); (B.Z.); (B.O.)
| |
Collapse
|
20
|
Bell S, Perkins GB, Anandh U, Coates PT. COVID and the Kidney: An Update. Semin Nephrol 2023; 43:151471. [PMID: 38199827 DOI: 10.1016/j.semnephrol.2023.151471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Coronavirus disease-2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2, has led to a global pandemic that continues to be responsible for ongoing health issues for people worldwide. Immunocompromised individuals such as kidney transplant recipients and dialysis patients have been and continue to be among the most affected, with poorer outcomes after infection, impaired response to COVID-19 vaccines, and protracted infection. The pandemic also has had a significant impact on patients with underlying chronic kidney disease (CKD), with CKD increasing susceptibility to COVID-19, risk of hospital admission, and mortality. COVID-19 also has been shown to lead to acute kidney injury (AKI) through both direct and indirect mechanisms. The incidence of COVID-19 AKI has been decreasing as the pandemic has evolved, but continues to be associated with adverse patient outcomes correlating with the severity of AKI. There is also increasing evidence examining the longer-term effect of COVID-19 on the kidney demonstrating continued decline in kidney function several months after infection. This review summarizes the current evidence examining the impact of COVID-19 on the kidney, covering both the impact on patients with CKD, including patients receiving kidney replacement therapy, in addition to discussing COVID-19 AKI.
Collapse
Affiliation(s)
- Samira Bell
- Division of Population Health and Genomics, University of Dundee, Dundee, Scotland.
| | - Griffith B Perkins
- University of Adelaide, South Australia, 5005 Australia; Central and Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide
| | - Urmila Anandh
- Department of Nephrology, Amrita Hospitals, Faridabad, Haryana, India
| | - P Toby Coates
- University of Adelaide, South Australia, 5005 Australia; Central and Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide
| |
Collapse
|
21
|
Vafea MT, Haidar G. COVID-19 Prevention in Solid Organ Transplant Recipients: Current State of the Evidence. Infect Dis Clin North Am 2023; 37:459-473. [PMID: 37217369 PMCID: PMC10030334 DOI: 10.1016/j.idc.2023.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Although COVID-19 vaccines are safe, most organ transplant recipients fail to mount an antibody response after two mRNA vaccines. Thus, three mRNA vaccines constitute a primary vaccine series after solid organ transplant. However, neutralizing antibodies after three or greater mRNA vaccines are lower against Omicron versus older variants. Predictors of attenuated responses include age, vaccination within 1 year from transplant, mycophenolate, and BNT162b2. Some seronegative transplant recipients exhibit durable T-cell responses. Vaccine effectiveness in transplants is lower than in the general population. Immunosuppression reduction around revaccination warrants further study. Monoclonal antibody pre-exposure prophylaxis may be protective against susceptible variants.
Collapse
Affiliation(s)
- Maria Tsikala Vafea
- Divison of Internal Medicine, University of Pittsburgh School of Medicine, UPMC, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Ghady Haidar
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, 3601 Fifth Avenue, Falk Medical Building, Suite 5B, Pittsburgh, PA 15213, USA.
| |
Collapse
|
22
|
Kuniduzi Y, Chen B, Zeng J, Sun X, Chen T, Qian X, Wang J, Liang F, Abuduxukuer R, Yusufu M, Xu S, Zhang X. Efficacy and safety of a fourth dose of the COVID-19 vaccine in kidney transplant recipients: A systematic review and meta-analysis. Transpl Immunol 2023; 79:101864. [PMID: 37230397 PMCID: PMC10205136 DOI: 10.1016/j.trim.2023.101864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/17/2023] [Accepted: 05/21/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND Kidney transplant recipients (KTRs) who become infected with SARS-CoV-2 are at greater risk of serious illness and death than the general population. To date, the efficacy and safety of the fourth dose of the COVID-19 vaccine in KTRs have not been systematically discussed. METHODS This systematic review and meta-analysis included articles from PubMed, Embase, the Cochrane Library, Web of Science, China National Knowledge Infrastructure, and Wanfang Med Online published before May 15, 2022. Studies evaluating the efficacy and safety of a fourth dose of the COVID-19 vaccine in kidney transplant recipients were selected. RESULTS Nine studies were included in the meta-analysis, with a total of 727 KTRs. The overall pooled seropositivity rate after the fourth COVID-19 vaccine was 60% (95% CI, 49%-71%, I2 = 87.83%, p > 0.01). The pooled proportion of KTRs seronegative after the third dose that transitioned to seropositivity after the fourth dose was 30% (95% CI, 15%-48%, I2 = 94.98%, p < 0.01). CONCLUSIONS The fourth dose of the COVID-19 vaccine was well tolerated in KTRs with no serious adverse effects. Some KTRs showed a reduced response even after receiving the fourth vaccine dose. Overall, the fourth vaccine dose effectively improved seropositivity in KTRs, as recommended by the World Health Organization for the general population.
Collapse
Affiliation(s)
- Yasen Kuniduzi
- College of Medicine, Wuhan University of Science and Technology, Wuhan, China; Center for Clinical Evidence-Based and Translational Medicine, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441021, China.; Department of Urology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Bo Chen
- Center for Clinical Evidence-Based and Translational Medicine, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441021, China
| | - Jingjing Zeng
- Center for Clinical Evidence-Based and Translational Medicine, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441021, China
| | - Xiaosong Sun
- Department of Urology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Tao Chen
- Department of Urology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Xiaoyuan Qian
- Department of Urology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Jiange Wang
- Department of Urology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Fuchao Liang
- Department of Urology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Rukeya Abuduxukuer
- College of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Maierhaba Yusufu
- College of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Shaoyong Xu
- Center for Clinical Evidence-Based and Translational Medicine, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441021, China.; Department of Endocrinology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441021, China.
| | - Xuejun Zhang
- Department of Urology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China.
| |
Collapse
|
23
|
Udomkarnjananun S, Gatechompol S, Leelahavanichkul A, Kerr SJ. Cellular immune response of SARS-CoV-2 vaccination in kidney transplant recipients: a systematic review and meta-analysis. Front Immunol 2023; 14:1220148. [PMID: 37575225 PMCID: PMC10415203 DOI: 10.3389/fimmu.2023.1220148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/05/2023] [Indexed: 08/15/2023] Open
Abstract
Background Evidence has demonstrated inferior humoral immune responses after SARS-CoV-2 vaccination in kidney transplant recipients compared to the general population. However, data on cellular immune responses in this population have not been established. Methods We searched the MEDLINE, Scopus, and Cochrane databases and included studies reporting cellular immune response rates in kidney transplant recipients after receiving SARS-CoV-2 vaccines. Studies that reported factors associated with cellular immune responders or non-responders were also included (PROSPERO: CRD42022375544). Results From a total of 1,494 articles searched, 53 articles were included in the meta-analysis. In all, 21 studies assessed cellular immune response by interferon-γ enzyme-linked immunosorbent spot (IFN-γ ELISPOT), 22 studies used interferon-γ release assay (IGRA), and 10 studies used flow cytometric analysis. The pooled response rate after two doses (standard regimen) and three doses of vaccination was 47.5% (95%CI 38.4-56.7%) and 69.1% (95%CI 56.3-80.6%) from IFN-γ ELISPOT, 25.8% (95%CI 19.7-32.4%) and 14.7% (95%CI 8.5-22.2%) from IGRA, and 73.7% (95%CI 55.2-88.8%) and 86.5% (95%CI 75.3-94.9%) from flow cytometry, respectively. Recipients with seroconversion were associated with a higher chance of having cellular immune response (OR 2.58; 95%CI 1.89-3.54). Cellular immune response in kidney transplant recipients was lower than in dialysis patients (OR 0.24; 95%CI 0.16-0.34) and the general population (OR 0.10; 95%CI 0.07-0.14). Age and immunosuppressants containing tacrolimus or corticosteroid were associated with inferior cellular immune response. Conclusion Cellular immune response after SARS-CoV-2 vaccination in kidney transplant recipients was lower than in dialysis patients and the general population. Age, tacrolimus, and corticosteroid were associated with poor response. Cellular immune response should also be prioritized in vaccination studies. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42022375544.
Collapse
Affiliation(s)
- Suwasin Udomkarnjananun
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
- Excellence Center for Organ Transplantation (ECOT), King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Renal Immunology and Transplantation Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Asada Leelahavanichkul
- Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Chulalongkorn University, Bangkok, Thailand
- Immunology Unit, Department of Microbiology, Chulalongkorn University, Bangkok, Thailand
| | - Stephen J. Kerr
- HIV-NAT, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
- Biostatistics Excellence Centre, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- The Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
24
|
Mu Y, Wu H, Jiang Z, Liu K, Xue X, Zhang W, Chen Z. Serological Responses after a Fourth Dose of SARS-CoV-2 Vaccine in Solid Organ Transplant Recipients: A Systematic Review and Meta-Analysis. Vaccines (Basel) 2023; 11:1130. [PMID: 37514946 PMCID: PMC10385971 DOI: 10.3390/vaccines11071130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 06/18/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
The humoral immune response and safety of the fourth dose of the coronavirus disease 2019 (COVID-19) vaccine in solid organ transplant (SOT) recipients need to be fully elucidated. We conducted a systematic review and meta-analysis to assess the efficacy and safety associated with this additional dose of the COVID-19 vaccine in the SOT recipients. A comprehensive search was conducted to identify studies on SOT patients without prior natural SARS-CoV-2 infection who received the fourth dose of the COVID-19 vaccine. Serological antibody responses following vaccination were synthesized by a meta-analysis of proportions. The proportions for each outcome were integrated by using a random-effects model. Approximately 56-92% of the SOT patients developed a humoral immune response, and the pooled seroprevalence rate was 75% (95% confidence interval [CI], 62-82%) after administering the third vaccine dose. Following the fourth dose of vaccination, approximately 76-95% of the patients developed a humoral immune response. The pooled seroprevalence rate after the fourth dose was 85% (95% CI, 79-91%). Of the patients who initially tested seronegative after the second dose, approximately 22-76% of patients subsequently became seropositive after the third dose. The pooled seroconversion rate for the third dose was 47% (95% CI, 31-64%). Among the patients who were seronegative after the third dose, approximately 25-76% turned seropositive after the fourth dose. The pooled seroconversion rate after the fourth dose was 51% (95% CI, 40-63%). Safety data were reported in three studies, demonstrating that adverse effects following the fourth dose were generally mild, and patients with these adverse effects did not require hospitalization. No transplant rejection or serious adverse events were observed. A fourth dose of the COVID-19 vaccine in SOT recipients was associated with an improved humoral immune response, and the vaccine was considered relatively safe.
Collapse
Affiliation(s)
- Yameng Mu
- Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing 100102, China
| | - Hongxiao Wu
- Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing 100102, China
| | - Zhouling Jiang
- Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing 100102, China
| | - Kehang Liu
- Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing 100102, China
| | - Xiaoyu Xue
- Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing 100102, China
| | - Wei Zhang
- Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing 100102, China
| | - Zhihai Chen
- Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing 100102, China
| |
Collapse
|
25
|
Emmanouilidou-Fotoulaki E, Karava V, Dotis J, Kondou A, Printza N. Immunologic Response to SARS-CoV-2 Vaccination in Pediatric Kidney Transplant Recipients: A Systematic Review and Meta-Analysis. Vaccines (Basel) 2023; 11:1080. [PMID: 37376469 DOI: 10.3390/vaccines11061080] [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: 03/26/2023] [Revised: 05/28/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
The pediatric population is at a lower risk of severe SARS-CoV-2 infection compared to adults. Nevertheless, immunosuppression in pediatric and adolescent kidney transplant recipients (KTRs) increases their hazard compared to the general population. This systematic review evaluates the efficacy of SARS-CoV-2 vaccines and determines the risk factors of no seroconversion in this population. PubMed-MEDLINE databases were searched for cohort studies. A meta-analysis was performed using fixed and random effect models. In total, seven studies including 254 patients were further analyzed. The random effect model demonstrated a 63% seroconversion rate (95% CI 0.5, 0.76) following a two-dose schedule, which increased to 85% (95% CI 0.76, 0.93) after the third dose administration. Seropositivity was lower in patients under mycophenolate mofetil compared to azathioprine (OR 0.09, 95% CI 0.02, 0.43). Rituximab administration decreased the seroconversion rate (OR 0.12, 95% CI 0.03, 0.43). The glomerular filtration rate (GFR) was 9.25 mL/min/1.73 m2 lower (95% CI 16.37, 2.13) in patients with no seroconversion. The seroconversion rate was lower in vaccinated compared to infected patients (OR 0.13, 95% CI 0.02, 0.72). In conclusion, vaccination against SARS-CoV-2 in pediatric and adolescent KTRs elicits a humoral response, and a third dose is advised. Previous rituximab administration, antimetabolite therapy with mycophenolate mofetil and lower GFR reduce the likelihood for seroconversion.
Collapse
Affiliation(s)
- Elpida Emmanouilidou-Fotoulaki
- Pediatric Nephrology Unit, 1st Department of Pediatrics, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| | - Vasiliki Karava
- Pediatric Nephrology Unit, 1st Department of Pediatrics, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| | - John Dotis
- Pediatric Nephrology Unit, 1st Department of Pediatrics, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| | - Antonia Kondou
- Pediatric Nephrology Unit, 1st Department of Pediatrics, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| | - Nikoleta Printza
- Pediatric Nephrology Unit, 1st Department of Pediatrics, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| |
Collapse
|
26
|
Rossi M, Pessolano G, Gambaro G. What has vaccination against COVID-19 in CKD patients taught us? J Nephrol 2023; 36:1257-1266. [PMID: 37140817 PMCID: PMC10157569 DOI: 10.1007/s40620-023-01640-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2023] [Indexed: 05/05/2023]
Abstract
Effective vaccination strategies are of crucial importance to protecting patients who are vulnerable to infections, such as patients with chronic kidney disease. This is because the decreased efficiency of the immune system in chronic kidney disease impairs vaccine-induced immunisation. COVID-19 has prompted investigation of the immune response to SARS-CoV-2 vaccines in chronic kidney disease and in kidney transplant recipients in an effort to improve efficacy. The seroconversion rate after two vaccine doses is reduced, especially in kidney transplant recipients. Furthermore, although the seroconversion rate in chronic kidney disease patients is as high as in healthy subjects, anti-spike antibody titres are lower than in healthy vaccinated individuals, and these titres decrease rapidly. Although the vaccine-induced anti-spike antibody titre correlates with neutralising antibody levels and with protection against COVID-19, the protective prognostic significance of their titre is decreased due to the emergence of SARS-CoV-2 variants other than the Wuhan index virus against which the original vaccines were produced. Cellular immunity is also relevant, and because of cross-reactivity to the spike protein, epitopes of different viral variants confer protection against newly emerging variants of SARS-CoV-2. A multi-dose vaccination strategy is the most effective way to obtain a sufficient serological response. In kidney transplant recipients, a 5-week discontinuation period from antimetabolite drugs in concomitance with vaccine administration may also increase the vaccine's efficacy. The newly acquired knowledge obtained from COVID-19 vaccination is of general interest for the success of other vaccinations in chronic kidney disease patients.
Collapse
Affiliation(s)
- Mattia Rossi
- Division of Nephrology, Department of Medicine, University of Verona, Piazzale A. Stefani 1, 37126, Verona, Italy.
| | - Giuseppina Pessolano
- Division of Nephrology, Department of Medicine, University of Verona, Piazzale A. Stefani 1, 37126, Verona, Italy
| | - Giovanni Gambaro
- Division of Nephrology, Department of Medicine, University of Verona, Piazzale A. Stefani 1, 37126, Verona, Italy
| |
Collapse
|
27
|
Werbel WA, Karaba AH, Chiang TPY, Massie AB, Brown DM, Watson N, Chahoud M, Thompson EA, Johnson AC, Avery RK, Cochran WV, Warren D, Liang T, Fribourg M, Huerta C, Samaha H, Klein SL, Bettinotti MP, Clarke WA, Sitaras I, Rouphael N, Cox AL, Bailey JR, Pekosz A, Tobian AAR, Durand CM, Bridges ND, Larsen CP, Heeger PS, Segev DL. Persistent SARS-CoV-2-specific immune defects in kidney transplant recipients following third mRNA vaccine dose. Am J Transplant 2023; 23:744-758. [PMID: 36966905 PMCID: PMC10037915 DOI: 10.1016/j.ajt.2023.03.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/17/2023] [Accepted: 03/17/2023] [Indexed: 03/26/2023]
Abstract
Kidney transplant recipients (KTRs) show poorer response to SARS-CoV-2 mRNA vaccination, yet response patterns and mechanistic drivers following third doses are ill-defined. We administered third monovalent mRNA vaccines to n = 81 KTRs with negative or low-titer anti-receptor binding domain (RBD) antibody (n = 39 anti-RBDNEG; n = 42 anti-RBDLO), compared with healthy controls (HCs, n = 19), measuring anti-RBD, Omicron neutralization, spike-specific CD8+%, and SARS-CoV-2-reactive T cell receptor (TCR) repertoires. By day 30, 44% anti-RBDNEG remained seronegative; 5% KTRs developed BA.5 neutralization (vs 68% HCs, P < .001). Day 30 spike-specific CD8+% was negative in 91% KTRs (vs 20% HCs; P = .07), without correlation to anti-RBD (rs = 0.17). Day 30 SARS-CoV-2-reactive TCR repertoires were detected in 52% KTRs vs 74% HCs (P = .11). Spike-specific CD4+ TCR expansion was similar between KTRs and HCs, yet KTR CD8+ TCR depth was 7.6-fold lower (P = .001). Global negative response was seen in 7% KTRs, associated with high-dose MMF (P = .037); 44% showed global positive response. Of the KTRs, 16% experienced breakthrough infections, with 2 hospitalizations; prebreakthrough variant neutralization was poor. Absent neutralizing and CD8+ responses in KTRs indicate vulnerability to COVID-19 despite 3-dose mRNA vaccination. Lack of neutralization despite CD4+ expansion suggests B cell dysfunction and/or ineffective T cell help. Development of more effective KTR vaccine strategies is critical. (NCT04969263).
Collapse
Affiliation(s)
- William A Werbel
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Andrew H Karaba
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Teresa Po-Yu Chiang
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Allan B Massie
- Department of Surgery, New York University Grossman School of Medicine, New York, New York, USA; Department of Population Health, New York University Grossman School of Medicine, New York, New York, USA
| | - Diane M Brown
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Natasha Watson
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Maggie Chahoud
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Elizabeth A Thompson
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Robin K Avery
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Willa V Cochran
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daniel Warren
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tao Liang
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Miguel Fribourg
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Hady Samaha
- Department of Medicine, Emory University, Atlanta, Georgia, USA
| | - Sabra L Klein
- Department of Medicine, Emory University, Atlanta, Georgia, USA
| | - Maria P Bettinotti
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - William A Clarke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ioannis Sitaras
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Nadine Rouphael
- Department of Medicine, Emory University, Atlanta, Georgia, USA
| | - Andrea L Cox
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Medicine, Emory University, Atlanta, Georgia, USA; Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Justin R Bailey
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Aaron A R Tobian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Christine M Durand
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nancy D Bridges
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Peter S Heeger
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Dorry L Segev
- Department of Surgery, New York University Grossman School of Medicine, New York, New York, USA; Department of Population Health, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
28
|
Poli MC, Vial C, Rey-Jurado E, González N, Cortés LJ, Hormazabal J, Ramírez-Riffo C, de la Cruz J, Ulloa C. A Third Dose of SARS-CoV-2 mRNA Vaccine Improves Immune Response in Chronic Kidney Disease Patients. Vaccines (Basel) 2023; 11:vaccines11051012. [PMID: 37243116 DOI: 10.3390/vaccines11051012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/28/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Chronic kidney disease (CKD) patients have an increased risk of morbidity and mortality following SARS-CoV-2 infection. Vaccination in these patients is prioritized, and monitoring of the immune response is paramount to define further vaccination strategies. This prospective study included a cohort of 100 adult CKD patients: 48 with kidney transplant (KT) and 52 on hemodialysis without prior COVID-19. The patients were assessed for humoral and cellular immune responses after four months of an anti-SARS-CoV-2 primary two-dose vaccination scheme (CoronaVac or BNT162b2) and one month after a booster third dose of BNT162b2 vaccine. We identified poor cellular and humoral immune responses in the CKD patients after a primary vaccination scheme, and these responses were improved by a booster. Robust polyfunctional CD4+ T cell responses were observed in the KT patients after a booster, and this could be attributed to a higher proportion of the patients having been vaccinated with homologous BNT162b2 schemes. However, even after the booster, the KT patients exhibited lower neutralizing antibodies, attributable to specific immunosuppressive treatments. Four patients suffered severe COVID-19 despite three-dose vaccination, and all had low polyfunctional T-cell responses, underscoring the importance of this functional subset in viral protection. In conclusion, a booster dose of SARS-CoV-2 mRNA vaccine in CKD patients improves the impaired humoral and cellular immune responses observed after a primary vaccination scheme.
Collapse
Affiliation(s)
- Maria Cecilia Poli
- Departamento de Pediatría, Clínica Alemana de Santiago, Santiago 7650568, Chile
- Programa de Inmunogenética e Inmunología Traslacional, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
| | - Cecilia Vial
- Programa Hantavirus y Zoonosis, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
| | - Emma Rey-Jurado
- Programa de Inmunogenética e Inmunología Traslacional, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
| | - Natalia González
- Programa de Inmunogenética e Inmunología Traslacional, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
- Programa Hantavirus y Zoonosis, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
| | - Lina Jimena Cortés
- Programa Hantavirus y Zoonosis, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
| | - Juan Hormazabal
- Programa Hantavirus y Zoonosis, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
| | - Carolina Ramírez-Riffo
- Programa Hantavirus y Zoonosis, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
| | - Javiera de la Cruz
- Programa de Inmunogenética e Inmunología Traslacional, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
| | - Camilo Ulloa
- Departamento de Medicina Interna, Unidad de Nefrología y Trasplante Renal, Clínica Alemana de Santiago, Santiago 7650568, Chile
| |
Collapse
|
29
|
Hausinger RI, Bachmann Q, Crone-Rawe T, Hannane N, Monsef I, Haller B, Heemann U, Skoetz N, Kreuzberger N, Schmaderer C. Effectiveness, Immunogenicity and Harms of Additional SARS-CoV-2 Vaccine Doses in Kidney Transplant Recipients: A Systematic Review. Vaccines (Basel) 2023; 11:vaccines11040863. [PMID: 37112775 PMCID: PMC10141039 DOI: 10.3390/vaccines11040863] [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: 02/20/2023] [Revised: 04/05/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Kidney transplant recipients (KTRs) who have a highly impaired immune response are in need of intensified and safe vaccination strategies to achieve seroconversion and prevent severe disease. METHODS We searched the Web of Science Core Collection, the Cochrane COVID-19 Study Register and the WHO COVID-19 global literature on coronavirus disease from January 2020 to 22 July 2022 for prospective studies that assessed immunogenicity and efficacy after three or more SARS-CoV-2 vaccine doses. RESULTS In 37 studies on 3429 patients, de novo seroconversion after three and four vaccine doses ranged from 32 to 60% and 25 to 37%. Variant-specific neutralization was 59 to 70% for Delta and 12 to 52% for Omicron. Severe disease after infection was rarely reported but all concerned KTRs lacked immune responses after vaccination. Studies investigating the clinical course of COVID-19 found remarkably higher rates of severe disease than in the general population. Serious adverse events and acute graft rejections were very rare. Substantial heterogeneity between the studies limited their comparability and summary. CONCLUSION Additional SARS-CoV-2 vaccine doses are potent and safe in general terms as well as regarding transplant-specific outcomes whilst the Omicron wave remains a significant threat to KTRs without adequate immune responses.
Collapse
Affiliation(s)
- Renate Ilona Hausinger
- Department of Nephrology, Klinikum Rechts der Isar, TUM School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Quirin Bachmann
- Department of Nephrology, Klinikum Rechts der Isar, TUM School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Timotius Crone-Rawe
- Department of Nephrology, Klinikum Rechts der Isar, TUM School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Nora Hannane
- Department of Nephrology, Klinikum Rechts der Isar, TUM School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Ina Monsef
- Evidence-Based Medicine, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Bernhard Haller
- Institute for AI and Informatics in Medicine, Klinikum Rechts der Isar, TUM School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Uwe Heemann
- Department of Nephrology, Klinikum Rechts der Isar, TUM School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Nicole Skoetz
- Evidence-Based Medicine, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Nina Kreuzberger
- Evidence-Based Medicine, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Christoph Schmaderer
- Department of Nephrology, Klinikum Rechts der Isar, TUM School of Medicine, Technical University of Munich, 81675 Munich, Germany
| |
Collapse
|
30
|
Martinelli S, Pascucci D, Laurenti P. Humoral response after a fourth dose of SARS-CoV-2 vaccine in immunocompromised patients. Results of a systematic review. Front Public Health 2023; 11:1108546. [PMID: 37033069 PMCID: PMC10076800 DOI: 10.3389/fpubh.2023.1108546] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
Background and objective The fourth dose the COVID-19 vaccine was first proposed to immunocompromised patients. The aim of the article is to systematically review the literature and report the humoral response and outcomes after the fourth dose administration in people with impaired immune system. Methods Published studies on the humoral response, efficacy and safety of the fourth dose of the COVID-19 vaccine were analyzed in various settings of immunocompromised patients. We conducted systematic searches of PubMed, Cochrane Library and WHO COVID-19 Research Database for series published through January 31, 2023, using the search terms "fourth dose" or "second booster" or "4th dose" and "Coronavirus" or "COVID-19" or "SARS-CoV-2." All articles were selected according to the PRISMA guidelines. Results A total of 24 articles including 2,838 patients were comprised in the systematic review. All the studies involved immunocompromised patients, including solid organ transplant recipients, patients with autoimmune rheumatic disease, patients with human immunodeficiency virus (HIV) and patients with blood cancers or diseases. Almost all patients received BNT162b2 or mRNA-1273 as fourth dose. All the studies demonstrated the increase of antibody titers after the fourth dose, both in patients who had a serological strong response and in those who had a weak response after the third dose. No serious adverse events after the 4th dose have been reported by 13 studies. COVID-19 infection after the fourth dose ranged from 0 to 21%. Conclusion The present review highlights the importance of the fourth dose of covid-19 vaccines for immunocompromised patients. Across the included studies, a fourth dose was associated with improved seroconversion and antibody titer levels. In particular, a fourth dose was associated with increasing immunogenicity in organ transplant recipients and patients with hematological cancers, with a very low rate of serious side effects.
Collapse
Affiliation(s)
- Silvia Martinelli
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Domenico Pascucci
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Patrizia Laurenti
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
31
|
SARS-CoV-2 vaccine strategies in kidney transplant recipients. THE LANCET. INFECTIOUS DISEASES 2023; 23:263-264. [PMID: 36354033 PMCID: PMC9612841 DOI: 10.1016/s1473-3099(22)00666-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022]
|
32
|
Kho MML, Messchendorp AL, Frölke SC, Imhof C, Koomen VJCH, Malahe SRK, Vart P, Geers D, de Vries RD, GeurtsvanKessel CH, Baan CC, van der Molen RG, Diavatopoulos DA, Remmerswaal EBM, van Baarle D, van Binnendijk R, den Hartog G, de Vries APJ, Gansevoort RT, Bemelman FJ, Reinders MEJ, Sanders JSF, Hilbrands LB, Baas MC, Bouwmans P, ten Dam MA, Gommers L, Standaar D, van der Heiden M, Adema YM, Boer-Verschragen MJ, Mattheussens WB, Philipsen RH, van Mourik D, Bogers S, van Dijk LL, Rots N, Smits G, Kuijer M, Hemmelder MH. Alternative strategies to increase the immunogenicity of COVID-19 vaccines in kidney transplant recipients not responding to two or three doses of an mRNA vaccine (RECOVAC): a randomised clinical trial. THE LANCET. INFECTIOUS DISEASES 2023; 23:307-319. [PMID: 36354032 PMCID: PMC9760034 DOI: 10.1016/s1473-3099(22)00650-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/11/2022] [Accepted: 09/12/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND An urgent need exists to improve the suboptimal COVID-19 vaccine response in kidney transplant recipients (KTRs). We aimed to compare three alternative strategies with a control single dose mRNA-1273 vaccination: a double vaccine dose, heterologous vaccination, and temporary discontinuation of mycophenolate mofetil or mycophenolic acid. METHODS This open-label randomised trial, done in four university medical centres in the Netherlands, enrolled KTRs without seroconversion after two or three doses of an mRNA vaccine. Between Oct 20, 2021, and Feb 2, 2022, 230 KTRs were randomly assigned block-wise per centre by a web-based system in a 1:1:1 manner to receive 100 μg mRNA-1273, 2 × 100 μg mRNA-1273, or Ad26.COV2-S vaccination. In addition, 103 KTRs receiving 100 μg mRNA-1273, were randomly assigned 1:1 to continue (mycophenolate mofetil+) or discontinue (mycophenolate mofetil-) mycophenolate mofetil or mycophenolic acid treatment for 2 weeks. The primary outcome was the percentage of participants with a spike protein (S1)-specific IgG concentration of at least 10 binding antibody units per mL at 28 days after vaccination, assessed in all participants who had a baseline measurement and who completed day 28 after vaccination without SARS-CoV-2 infection. Safety was assessed as a secondary outcome in all vaccinated patients by incidence of solicited adverse events, acute rejection or other serious adverse events. This trial is registered with ClinicalTrials.gov, NCT05030974 and is closed. FINDINGS Between April 23, 2021, and July 2, 2021, of 12 158 invited Dutch KTRs, 3828 with a functioning kidney transplant participated in a national survey for antibody measurement after COVID-19 vaccination. Of these patients, 1311 did not seroconvert after their second vaccination and another 761 not even after a third. From these seronegative patients, 345 agreed to participate in our repeated vaccination study. Vaccination with 2 × mRNA-1273 or Ad26.COV2-S was not superior to single mRNA-1273, with seroresponse rates of 49 (68%) of 72 (95% CI 56-79), 46 (63%) of 73 (51-74), and 50 (68%) of 73 (57-79), respectively. The difference with single mRNA-1273 was -0·4% (-16 to 15; p=0·96) for 2 × mRNA-1273 and -6% (-21 to 10; p=0·49) for Ad26.COV2-S. Mycophenolate mofetil- was also not superior to mycophenolate mofetil+, with seroresponse rates of 37 (80%) of 46 (66-91) and 31 (67%) of 46 (52-80), and a difference of 13% (-5 to 31; p=0·15). Local adverse events were more frequent after a single and double dose of mRNA-1273 than after Ad26.COV2-S (65 [92%] of 71, 67 [92%] of 73, and 38 [50%] of 76, respectively; p<0·0001). No acute rejection occurred. There were no serious adverse events related to vaccination. INTERPRETATION Repeated vaccination increases SARS-CoV-2-specific antibodies in KTRs, without further enhancement by use of a higher dose, a heterologous vaccine, or 2 weeks discontinuation of mycophenolate mofetil or mycophenolic acid. To achieve a stronger response, possibly required to neutralise new virus variants, repeated booster vaccination is needed. FUNDING The Netherlands Organization for Health Research and Development and the Dutch Kidney Foundation.
Collapse
Affiliation(s)
- Marcia M L Kho
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus Medical Center, Rotterdam, Netherlands
| | - A Lianne Messchendorp
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Sophie C Frölke
- Renal Transplant Unit, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Celine Imhof
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands,Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Vera JCH Koomen
- Department of Nephrology, Radboud University Medical Center Nijmegen, Nijmegen, Netherlands
| | - S Reshwan K Malahe
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus Medical Center, Rotterdam, Netherlands
| | - Priya Vart
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Daryl Geers
- Department Viroscience, Erasmus MC Transplant Institute, Erasmus Medical Center, Rotterdam, Netherlands
| | - Rory D de Vries
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus Medical Center, Rotterdam, Netherlands
| | - Corine H GeurtsvanKessel
- Department Viroscience, Erasmus MC Transplant Institute, Erasmus Medical Center, Rotterdam, Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus Medical Center, Rotterdam, Netherlands
| | - Renate G van der Molen
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center Nijmegen, Nijmegen, Netherlands
| | - Dimitri A Diavatopoulos
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center Nijmegen, Nijmegen, Netherlands,Radboud Center for Infectious Diseases, Radboud University Medical Center Nijmegen, Nijmegen, Netherlands
| | - Ester B M Remmerswaal
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Debbie van Baarle
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, Netherlands,Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Rob van Binnendijk
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Gerco den Hartog
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Aiko P J de Vries
- Department Viroscience, Erasmus MC Transplant Institute, Erasmus Medical Center, Rotterdam, Netherlands,Department of Nephrology, Leiden University Medical Center, Leiden, Netherlands
| | - Ron T Gansevoort
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Frederike J Bemelman
- Renal Transplant Unit, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Marlies E J Reinders
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus Medical Center, Rotterdam, Netherlands
| | - Jan-Stephan F Sanders
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Luuk B Hilbrands
- Department of Nephrology, Radboud University Medical Center Nijmegen, Nijmegen, Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Meziyerh S, Bouwmans P, van Gelder T, van der Helm D, Messchendorp L, van der Boog PJM, de Fijter JW, Moes DJAR, de Vries APJ. Mycophenolic Acid Exposure Determines Antibody Formation Following SARS-CoV-2 Vaccination in Kidney Transplant Recipients: A Nested Cohort Study. Clin Pharmacol Ther 2023. [PMID: 36789469 DOI: 10.1002/cpt.2872] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/07/2023] [Indexed: 02/16/2023]
Abstract
Despite (repeated) boosting, kidney transplant recipients (KTRs) may remain at increased risk of severe COVID-19 since a substantial number of individuals remain seronegative or with low antibody titers. In particular, mycophenolic acid use has been shown to affect antibody formation negatively and may be an important modifiable risk factor. We investigated the exposure-response relationship between mycophenolic acid 12-hour area under the curve (AUC0-12h ) exposure and seroconversion including antibody titers after vaccination using mRNA-1273 SARS-CoV-2 vaccine (Moderna) in 316 KTRs from our center that participated in the national Dutch renal patients COVID-19 vaccination - long term efficacy and safety of SARS-CoV-2 vaccination in kidney disease patients vaccination study. After two vaccination doses, 162 (51%) KTRs seroconverted. KTRs treated with mycophenolic acid showed less seroconversion and lower antibody titers compared with KTRs without mycophenolic acid (44% vs. 77%, and 36 binding antibody units (BAU)/mL vs. 340 BAU/mL; P < 0.001). The mean mycophenolic acid AUC0-12h exposure was significantly lower in KTRs who seroconverted compared with KTRs who did not (39 vs. 29 mg⋅h/L; P < 0.001). High mycophenolic acid exposure (±90 mg⋅h/L) and no exposure to mycophenolic acid resulted in a seroconversion rate ranging from 10% to 80%. Every 10 mg⋅h/L increase in mycophenolic acid AUC0-12h gave an adjusted odds ratio for seroconversion of 0.87 (95% confidence interval (CI), 0.79-0.97; P = 0.010) and 0.89 (95% CI, 0.85-0.93; P < 0.001) for KTRs on dual and triple maintenance immunosuppressive therapy, respectively. Higher mycophenolic acid AUC0-12h correlated with lower antibody titers (R = 0.44, P < 0.001). This study demonstrates the exposure-response relationship between gold standard mycophenolic acid exposure and antibody formation to support interventional studies investigating mycophenolic acid adjustment to improve antibody formation after further boosting.
Collapse
Affiliation(s)
- Soufian Meziyerh
- Department of Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden University Medical Center Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Pim Bouwmans
- Department of Internal Medicine, Division of Nephrology, Maastricht University Medical Center, Maastricht, The Netherlands.,Cardiovascular Research Institute Maastricht School for Cardiovascular Disease, University of Maastricht, Maastricht, The Netherlands
| | - Teun van Gelder
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Danny van der Helm
- Leiden University Medical Center Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Lianne Messchendorp
- Department of Nephrology, University Medical Center Groningen, Groningen, The Netherlands
| | - Paul J M van der Boog
- Department of Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden University Medical Center Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Johan W de Fijter
- Department of Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden University Medical Center Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Dirk Jan A R Moes
- Cardiovascular Research Institute Maastricht School for Cardiovascular Disease, University of Maastricht, Maastricht, The Netherlands
| | - Aiko P J de Vries
- Department of Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden University Medical Center Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
34
|
Baran DA, Kapoor S, Grewal J, Andries G, Camacho M. Long term follow-up of the tacrolimus in combination, tacrolimus alone compared (TICTAC) trial. J Heart Lung Transplant 2023; 42:838-845. [PMID: 36870863 DOI: 10.1016/j.healun.2023.01.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 01/15/2023] [Accepted: 01/26/2023] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND Tacrolimus (TAC) monotherapy has been compared to TAC and mycophenolate mofetil (MMF) in the randomized Tacrolimus in Combination, Tacrolimus Alone Compared (TICTAC) trial. Long term results are now reported. METHODS Demographics are presented with descriptive statistics. Time to event was determined with Kaplan-Meier plots and Mantel-Cox Logrank statistics used to compare groups. RESULTS One hundred and forty-seven (98 %) of the initial 150 TICTAC trial patients had long-term follow-up data available. The median follow-up was 13.4 years (interquartile range 7.2-15.1 years). Post-transplant survival at 5, 10 and 15 years in the TAC monotherapy group was 84.5 %, 66.9 %, and 52.7 %, and 94.4 %, 78.2 % and 56.1 % for patients randomized to TAC / MMF (p = 0.19 logrank). The freedom from cardiac allograft vasculopathy (≥grade 1) was 100 %, 87.5 %, 69.3 % and 46.5 % at 1, 5, 10 and 15 years in the monotherapy group and 100 %, 76.9 %, 68.1 % and 54.4 % in the TAC/MMF group respectively (p = 0.96 logrank). Crossover of treatment assignment did not alter these findings. The freedom from dialysis or renal replacement was 92.8 %, 84.2 % and 68.4 % for TAC monotherapy patients versus 100 %, 93.4 % and 82.3 % for TAC/MMF patients at 5, 10 and 15-years post-transplant (p = 0.15 logrank). CONCLUSIONS Patients randomized to TAC/ MMF with 8-week steroid weaning had comparable outcomes to those with similar steroid regimen but discontinuation of MMF at 2 week post-transplant. The best outcomes were noted for patients initiated on TAC/ MMF including those where MMF was discontinued for intolerance. Both strategies are reasonable alternatives for patients post heart transplant. CONDENSED ABSTRACT Tacrolimus monotherapy was compared to TAC and mycophenolate mofetil without long term steroids in the randomized Tacrolimus in Combination, Tacrolimus Alone Compared (TICTAC) trial. Post-transplant survival at 5, 10 and 15 years in the TAC monotherapy group was 84.5%, 66.9 %, and 52.7 %, and 94.4 %, 78.2 % and 56.1 % for patients randomized to TAC / MMF (p = 0.19 logrank). Cardiac allograft vasculopathy and kidney failure were similar between groups. Immunosuppression should be individualized to avoid over treating some patients while undertreating others.
Collapse
Affiliation(s)
- David A Baran
- Department of Cardiology, Cleveland Clinic Heart, Vascular and Thoracic Institute, Cleveland, Ohio.
| | - Saurabh Kapoor
- Division of Cardiology, Newark Beth Israel Medical Center, Heart Failure Treatment and Transplant Program, Newark, New Jersey
| | - Jagpreet Grewal
- Division of Cardiology, Newark Beth Israel Medical Center, Heart Failure Treatment and Transplant Program, Newark, New Jersey
| | - Gabriela Andries
- Division of Cardiology, Newark Beth Israel Medical Center, Heart Failure Treatment and Transplant Program, Newark, New Jersey
| | - Margarita Camacho
- Division of Cardiothoracic Surgery, Newark Beth Israel Medical Center, Newark, New Jersey
| |
Collapse
|
35
|
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: 4.0] [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
|
36
|
Donor-Derived Cell-free DNA for Personalized Immunosuppression in Renal Transplantation. Ther Drug Monit 2023; 45:20-25. [PMID: 36127770 DOI: 10.1097/ftd.0000000000001023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/13/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND The long-term outcomes of solid organ transplantation remain suboptimal. Therefore, appropriate biomarkers are needed in addition to immunosuppressive drugs and other traditional approaches for graft monitoring to achieve personalized immunosuppression and reduce premature graft loss. METHODS Donor-derived cell-free DNA (dd-cfDNA) is a minimally invasive biomarker of cell death due to graft injury. It can be quantified using droplet digital polymerase chain reaction and next-generation sequencing. Fractional dd-cfDNA determination can be affected by changes in recipient cfDNA, such as those caused by leukopenia or infection, leading to false-positive or false-negative results, respectively. Absolute quantification of dd-cfDNA helps in overcoming this limitation. RESULTS Overall, there is sufficient evidence of the clinical validity of dd-cfDNA. It detects rejection episodes early at an actionable stage and reflects the severity of graft injury without being rejection-specific. Owing to its high negative predictive value, dd-cfDNA is very useful for ruling out graft injury. Dd-cfDNA complements histological findings and can help in avoiding unnecessary biopsies. It indicates a response to rejection treatment and detects underimmunosuppression. CONCLUSIONS Monitoring changes in dd-cfDNA over time may be helpful in adapting immunosuppression to prevent graft rejection. Moreover, serial dd-cfDNA determination may increase the effectiveness of transplant recipient surveillance and facilitate personalized immunosuppression when combined with other relevant clinical and diagnostic findings.
Collapse
|
37
|
Immune Response to COVID-19 mRNA Vaccination in Previous Nonresponder Kidney Transplant Recipients After Short-term Withdrawal of Mycophenolic Acid 1 and 3 Months After an Additional Vaccine Dose. Transplantation 2023; 107:1139-1150. [PMID: 36617671 PMCID: PMC10125015 DOI: 10.1097/tp.0000000000004516] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND The impaired immune response to coronavirus disease 2019 (COVID-19) vaccination in kidney transplant recipients (KTRs) leads to an urgent need for adapted immunization strategies. METHODS Sixty-nine KTRs without seroconversion after ≥3 COVID-19 vaccinations were enrolled, and humoral response was determined after an additional full-dose mRNA-1273 vaccination by measuring severe acute respiratory syndrome coronavirus 2-specific antibodies and neutralizing antibody activity against the Delta and Omicron variants 1 and 3 mo postvaccination. T-cell response was analyzed 3 mo postvaccination by assessing interferon-γ release. Mycophenolic acid (MPA) was withdrawn in 41 KTRs 1 wk before until 4 wk after vaccination to evaluate effects on immunogenicity. Graft function, changes in donor-specific anti-HLA antibodies, and donor-derived cell-free DNA were monitored in KTRs undergoing MPA withdrawal. RESULTS Humoral response to vaccination was significantly stronger in KTRs undergoing MPA withdrawal 1 mo postvaccination; however, overall waning humoral immunity was noted in all KTRs 3 mo after vaccination. Higher anti-S1 immunoglobulin G levels correlated with better neutralizing antibody activity against the Delta and Omicron variants, whereas no significant association was detected between T-cell response and neutralizing antibody activity. No rejection occurred during study, and graft function remained stable in KTRs undergoing MPA withdrawal. In 22 KTRs with Omicron variant breakthrough infections, neutralizing antibody activity was better against severe acute respiratory syndrome coronavirus 2 wild-type and the Delta variants than against the Omicron variant. CONCLUSIONS MPA withdrawal to improve vaccine responsiveness should be critically evaluated because withdrawing MPA may be associated with enhanced alloimmune response, and the initial effect of enhanced seroconversion rates in KTRs with MPA withdrawal disappears 3 mo after vaccination.
Collapse
|
38
|
Vinke JSJ, Altulea DHA, Eisenga MF, Jagersma RL, Niekolaas TM, van Baarle D, Heiden MVD, Steenhuis M, Rispens T, Abdulahad WH, Sanders JSF, De Borst MH. Ferric carboxymaltose and SARS-CoV-2 vaccination-induced immunogenicity in kidney transplant recipients with iron deficiency: The COVAC-EFFECT randomized controlled trial. Front Immunol 2023; 13:1017178. [PMID: 36618359 PMCID: PMC9822258 DOI: 10.3389/fimmu.2022.1017178] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 12/05/2022] [Indexed: 12/25/2022] Open
Abstract
Background Kidney transplant recipients (KTRs) have an impaired immune response after vaccination against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Iron deficiency (ID) may adversely affect immunity and vaccine efficacy. We aimed to investigate whether ferric carboxymaltose (FCM) treatment improves humoral and cellular responses after SARS-CoV-2 vaccination in iron-deficient KTRs. Methods We randomly assigned 48 iron-deficient KTRs to intravenous FCM (1-4 doses of 500mg with six-week intervals) or placebo. Co-primary endpoints were SARS-CoV-2-specific anti-Receptor Binding Domain (RBD) Immunoglobulin G (IgG) titers and T-lymphocyte reactivity against SARS-CoV-2 at four weeks after the second vaccination with mRNA-1273 or mRNA-BNT162b2. Results At four weeks after the second vaccination, patients receiving FCM had higher plasma ferritin and transferrin saturation (P<0.001 vs. placebo) and iron (P=0.02). However, SARS-CoV-2-specific anti-RBD IgG titers (FCM: 66.51 [12.02-517.59] BAU/mL; placebo: 115.97 [68.86-974.67] BAU/mL, P=0.07) and SARS-CoV-2-specific T-lymphocyte activation (FCM: 93.3 [0.85-342.5] IFN-ɣ spots per 106 peripheral blood mononuclear cells (PBMCs), placebo: 138.3 [0.0-391.7] IFN-ɣ spots per 106 PBMCs, P=0.83) were not significantly different among both arms. After the third vaccination, SARS-CoV-2-specific anti-RBD IgG titers remained similar between treatment groups (P=0.99). Conclusions Intravenous iron supplementation efficiently restored iron status but did not improve the humoral or cellular immune response against SARS-CoV-2 after three vaccinations.
Collapse
Affiliation(s)
| | - Dania H. A. Altulea
- Department of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| | - Michele F. Eisenga
- Department of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| | - Renate L. Jagersma
- Department of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| | - Tessa M. Niekolaas
- Department of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| | - Debbie van Baarle
- Department of Immunology, University Medical Center Groningen, Groningen, Netherlands
| | | | - Maurice Steenhuis
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
| | - Wayel H. Abdulahad
- Department of Immunology, University Medical Center Groningen, Groningen, Netherlands
| | | | - Martin H. De Borst
- Department of Nephrology, University Medical Center Groningen, Groningen, Netherlands,*Correspondence: Martin H. De Borst,
| |
Collapse
|
39
|
O’Callaghan JM. Transplant Trial Watch. Transpl Int 2023; 36:11202. [PMID: 37025500 PMCID: PMC10070464 DOI: 10.3389/ti.2023.11202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 02/01/2023] [Indexed: 04/08/2023]
Affiliation(s)
- John Matthew O’Callaghan
- University Hospitals Coventry and Warwickshire, Coventry, United Kingdom
- Centre for Evidence in Transplantation, University of Oxford, Oxford, United Kingdom
- *Correspondence: John Matthew O’Callaghan,
| |
Collapse
|
40
|
Kim HJ, Lee HJ, Yu S, Shin KH, Cho WH, Yeo HJ. Immunosuppressant and SARS-CoV-2 Vaccine Antibody Response After Lung Transplantation. Transplant Proc 2022; 54:2692-2697. [PMID: 36411096 PMCID: PMC9663737 DOI: 10.1016/j.transproceed.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND There are insufficient reports on the immunogenicity and safety of the COVID-19 vaccination after lung transplantation in Korea. METHODS Between April and September 2021, lung transplant recipients (n = 52) and healthy controls (n = 22) underwent vaccination. The levels of antibodies were assessed prospectively at 4 weeks after priming and second dose. RESULTS Of a total of 52 lung transplant recipients, there were 84.6% nonresponders, 15.4% second-dose responders, and 0% primary dose responders. Among healthy controls, 63.6% were priming responders, and 18.2% were second-dose responders, and 18.2% were nonresponders. Compared with the control group, lung recipients were less likely to develop antibodies (P < .001). Antibody formation tended to be higher in recipients more than 1 year after transplantation (0 vs 20.5%, P = .076). No major safety events were reported, and the adverse symptoms were mild and consistent with those of the general population. In a multivariate regression analysis, mycophenolic acid levels (µg/mL) (odds ratio 0.25, P = .005) and tacrolimus level (ng/mL) (odds ratio 0.65, P = .035) were significantly associated with antibody formation. CONCLUSIONS The immunogenicity of the second dose of COVID-19 vaccination with various combinations was substantially low in lung transplants. A booster of the COVID-19 vaccine is warranted in lung transplants, especially a year later.
Collapse
Affiliation(s)
- Hye-Jin Kim
- Department of Anesthesia and Pain Medicine, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Hyun Ji Lee
- Department of Laboratory Medicine, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Shinae Yu
- Department of Laboratory Medicine, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Kyung-Hwa Shin
- Department of Laboratory Medicine, Pusan National University School of Medicine, and Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Woo Hyun Cho
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Hye Ju Yeo
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea,Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea,Address correspondence to Hye Ju Yeo, MD, PhD, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, 20 Geumo-ro, Mulgeumeup, Yangsan 50612, Republic of Korea
| |
Collapse
|
41
|
Bouwmans P, Messchendorp AL, Imhof C, Sanders JSF, Hilbrands LB, Reinders MEJ, Vart P, Bemelman FJ, Abrahams AC, van den Dorpel RMA, Ten Dam MAGJ, de Vries APJ, Rispens T, Steenhuis M, Gansevoort RT, Hemmelder MH. Impact of immunosuppressive treatment and type of SARS-CoV-2 vaccine on antibody levels after three vaccinations in patients with chronic kidney disease or kidney replacement therapy. Clin Kidney J 2022; 16:528-540. [PMID: 36865021 PMCID: PMC9972832 DOI: 10.1093/ckj/sfac249] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Indexed: 11/26/2022] Open
Abstract
Background Patients with chronic kidney disease (CKD) or kidney replacement therapy demonstrate lower antibody levels after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination compared with healthy controls. In a prospective cohort, we analysed the impact of immunosuppressive treatment and type of vaccine on antibody levels after three SARS-CoV-2 vaccinations. Methods Control subjects (n = 186), patients with CKD G4/5 (n = 400), dialysis patients (n = 480) and kidney transplant recipients (KTR) (n = 2468) were vaccinated with either mRNA-1273 (Moderna), BNT162b2 (Pfizer-BioNTech) or AZD1222 (Oxford/AstraZeneca) in the Dutch SARS-CoV-2 vaccination programme. Third vaccination data were available in a subgroup of patients (n = 1829). Blood samples and questionnaires were obtained 1 month after the second and third vaccination. Primary endpoint was the antibody level in relation to immunosuppressive treatment and type of vaccine. Secondary endpoint was occurrence of adverse events after vaccination. Results Antibody levels after two and three vaccinations were lower in patients with CKD G4/5 and dialysis patients with immunosuppressive treatment compared with patients without immunosuppressive treatment. After two vaccinations, we observed lower antibody levels in KTR using mycophenolate mofetil (MMF) compared with KTR not using MMF [20 binding antibody unit (BAU)/mL (3-113) vs 340 BAU/mL (50-1492), P < .001]. Seroconversion was observed in 35% of KTR using MMF, compared with 75% of KTR not using MMF. Of the KTR who used MMF and did not seroconvert, eventually 46% seroconverted after a third vaccination. mRNA-1273 induces higher antibody levels as well as a higher frequency of adverse events compared with BNT162b2 in all patient groups. Conclusions Immunosuppressive treatment adversely affects the antibody levels after SARS-CoV-2 vaccination in patients with CKD G4/5, dialysis patients and KTR. mRNA-1273 vaccine induces a higher antibody level and higher frequency of adverse events.
Collapse
Affiliation(s)
- Pim Bouwmans
- Department of Internal Medicine, Division of Nephrology, Maastricht University Medical Center, Maastricht, The Netherlands
- CARIM School for Cardiovascular Diseases, University of Maastricht, Maastricht, The Netherlands
| | - A Lianne Messchendorp
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Céline Imhof
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jan-Stephan F Sanders
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Luuk B Hilbrands
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marlies E J Reinders
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Priya Vart
- Department of Internal Medicine, University Medical Center Groningen, Groningen, The Netherlands
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, The Netherlands
| | - Frederike J Bemelman
- Amsterdam UMC Location University of Amsterdam, Renal Transplant Unit, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands
| | - Alferso C Abrahams
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Marc A G J Ten Dam
- Department of Internal Medicine, Canisius Wilhelmina Hospital, Nijmegen, The Netherlands
| | - Aiko P J de Vries
- Department of Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
- Leiden Transplant Center, Leiden, The Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research, Amsterdam, The Netherlands
- Landsteiner Laboratory, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Maurice Steenhuis
- Department of Immunopathology, Sanquin Research, Amsterdam, The Netherlands
- Landsteiner Laboratory, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Ron T Gansevoort
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | |
Collapse
|
42
|
Kodali L, Budhiraja P, Gea-Banacloche J. COVID-19 in kidney transplantation-implications for immunosuppression and vaccination. Front Med (Lausanne) 2022; 9:1060265. [PMID: 36507509 PMCID: PMC9727141 DOI: 10.3389/fmed.2022.1060265] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2022] Open
Abstract
COVID-19 pandemic continues to challenge the transplant community, given increased morbidity and mortality associated with the disease and poor response to prevention measures such as vaccination. Transplant recipients have a diminished response to both mRNA and vector-based vaccines compared to dialysis and the general population. The currently available assays to measure response to vaccination includes commercially available antibody assays for anti-Spike Ab, or anti- Receptor Binding Domain Ab. Positive antibody testing on the assays does not always correlate with neutralizing antibodies unless the antibody levels are high. Vaccinations help with boosting polyfunctional CD4+ T cell response, which continues to improve with subsequent booster doses. Ongoing efforts to improve vaccine response by using additional booster doses and heterologous vaccine combinations are underway. There is improved antibody response in moderate responders; however, the ones with poor response to initial vaccination doses, continue to have a poor response to sequential boosters. Factors associated with poor vaccine response include diabetes, older age, specific immunosuppressants such as belatacept, and high dose mycophenolate. In poor responders, a decrease in immunosuppression can increase response to vaccination. COVID infection or vaccination has not been associated with an increased risk of rejection. Pre- and Post-exposure monoclonal antibodies are available to provide further protection against COVID infection, especially in poor vaccine responders. However, the efficacy is challenged by the emergence of new viral strains. A recently approved bivalent vaccine offers better protection against the Omicron variant.
Collapse
Affiliation(s)
- Lavanya Kodali
- Department of Internal Medicine, Mayo Clinic, Phoenix, AZ, United States
- Division of Nephrology, Transplant Center, Mayo Clinic, Phoenix, AZ, United States
| | - Pooja Budhiraja
- Department of Internal Medicine, Mayo Clinic, Phoenix, AZ, United States
- Division of Nephrology, Transplant Center, Mayo Clinic, Phoenix, AZ, United States
| | - Juan Gea-Banacloche
- Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| |
Collapse
|
43
|
Stich M, Di Cristanziano V, Tönshoff B, Weber LT, Dötsch J, Rammer MT, Rieger S, Heger E, Garbade SF, Burgmaier K, Benning L, Speer C, Habbig S, Haumann S. Humoral immune response and live-virus neutralization of the SARS-CoV-2 omicron (BA.1) variant after COVID-19 mRNA vaccination in children and young adults with chronic kidney disease. Pediatr Nephrol 2022; 38:1935-1948. [PMID: 36409368 PMCID: PMC9684918 DOI: 10.1007/s00467-022-05806-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Data on humoral immune response to standard COVID-19 vaccination are scarce in adolescent patients and lacking for children below 12 years of age with chronic kidney disease including kidney transplant recipients. METHODS We therefore investigated in this retrospective two-center study (DRKS00024668; registered 23.03.2021) the humoral immune response to a standard two-dose mRNA vaccine regimen in 123 CKD patients aged 5-30 years. A live-virus assay was used to assess the serum neutralizing activity against the SARS-CoV-2 omicron (BA.1) variant. RESULTS Children aged 5-11 years had a comparable rate and degree of immune response to adolescents despite lower vaccine doses (10 µg vs. 30 µg BNT162b2). Treatment with two (odds ratio 9.24) or three or more (odds ratio 17.07) immunosuppressants was an independent risk factor for nonresponse. The immune response differed significantly among three patient cohorts: 48 of 77 (62.3%) kidney transplant recipients, 21 of 26 (80.8%) patients on immunosuppressive therapy, and 19 of 20 (95.0%) patients with chronic kidney disease without immunosuppressive therapy responded. In the kidney transplant recipients, immunosuppressive regimens comprising mycophenolate mofetil, an eGFR of < 60 mL/min/1.73 m2, and female sex were independent risk factors for nonresponse. Two of 18 (11.1%) and 8 of 16 (50.0%) patients with an anti-S1-RBD IgG of 100-1411 and > 1411 BAU/mL, respectively, showed a neutralization activity against the omicron variant. CONCLUSION A standard mRNA vaccine regimen in immunosuppressed children and adolescents with kidney disease elicits an attenuated humoral immune response with effective live virus neutralization against the omicron variant in approximately 10% of the patients, underlying the need for omicron-adapted vaccination. A higher resolution version of the Graphical abstract is available as Supplementary information.
Collapse
Affiliation(s)
- Maximilian Stich
- Department of Pediatrics I, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Veronica Di Cristanziano
- Institute of Virology, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Burkhard Tönshoff
- Department of Pediatrics I, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Lutz Thorsten Weber
- Department of Pediatrics, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Jörg Dötsch
- Department of Pediatrics, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Marian Theodor Rammer
- Department of Pediatrics, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Susanne Rieger
- Department of Pediatrics I, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Eva Heger
- Institute of Virology, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Sven F Garbade
- Department of Pediatrics I, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Kathrin Burgmaier
- Department of Pediatrics, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
- Faculty of Applied Healthcare Science, Deggendorf Institute of Technology, Deggendorf, Germany
| | - Louise Benning
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - Claudius Speer
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - Sandra Habbig
- Department of Pediatrics, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany.
| | - Sophie Haumann
- Department of Pediatrics, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
| |
Collapse
|
44
|
Thomson T, Prendecki M, Gleeson S, Martin P, Spensley K, De Aguiar RC, Sandhu B, Seneschall C, Gan J, Clarke CL, Lewis S, Pickard G, Thomas D, McAdoo SP, Lightstone L, Cox A, Kelleher P, Willicombe M. Immune responses following 3rd and 4th doses of heterologous and homologous COVID-19 vaccines in kidney transplant recipients. EClinicalMedicine 2022; 53:101642. [PMID: 36105874 PMCID: PMC9462844 DOI: 10.1016/j.eclinm.2022.101642] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Solid organ transplant recipients have attenuated immune responses to SARS-CoV-2 vaccines. In this study, we report on immune responses to 3rd- (V3) and 4th- (V4) doses of heterologous and homologous vaccines in a kidney transplant population. METHODS We undertook a single centre cohort study of 724 kidney transplant recipients prospectively screened for serological responses following 3 primary doses of a SARS-CoV2 vaccine. 322 patients were sampled post-V4 for anti-spike (anti-S), with 69 undergoing assessment of SARS-CoV-2 T-cell responses. All vaccine doses were received post-transplant, only mRNA vaccines were used for V3 and V4 dosing. All participants had serological testing performed post-V2 and at least once prior to their first dose of vaccine. FINDINGS 586/724 (80.9%) patients were infection-naïve post-V3; 141/2586 (24.1%) remained seronegative at 31 (21-51) days post-V3. Timing of vaccination in relation to transplantation, OR: 0.28 (0.15-0.54), p=0.0001; immunosuppression burden, OR: 0.22 (0.13-0.37), p<0.0001, and a diagnosis of diabetes, OR: 0.49 (0.32-0.75), p=0.001, remained independent risk factors for non-seroconversion. Seropositive patients post-V3 had greater anti-S if primed with BNT162b2 compared with ChAdOx1, p=0.001.Post-V4, 45/239 (18.8%) infection-naïve patients remained seronegative. De novo seroconversion post-V4 occurred in 15/60 (25.0%) patients. There was no difference in anti-S post-V4 by vaccine combination, p=0.50. T-cell responses were poor, with only 11/54 (20.4%) infection-naive patients having detectable T-cell responses post-V4, with no difference seen by vaccine type. INTERPRETATION A significant proportion of transplant recipients remain seronegative following 3- and 4- doses of SARS-CoV-2 vaccines, with poor T-cell responses, and are likely to have inadequate protection against infection. As such alternative strategies are required to provide protection to this vulnerable group. FUNDING MW/PK received study support from Oxford Immunotec.
Collapse
Affiliation(s)
- Tina Thomson
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, W12 0HS, United Kingdom
| | - Maria Prendecki
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, W12 0HS, United Kingdom
| | - Sarah Gleeson
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, W12 0HS, United Kingdom
| | - Paul Martin
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, W12 0HS, United Kingdom
| | - Katrina Spensley
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, W12 0HS, United Kingdom
| | - Rute Cardoso De Aguiar
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, W12 0HS, United Kingdom
| | - Bynvant Sandhu
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, W12 0HS, United Kingdom
| | - Charlotte Seneschall
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, W12 0HS, United Kingdom
| | - Jaslyn Gan
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, W12 0HS, United Kingdom
| | - Candice L. Clarke
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, W12 0HS, United Kingdom
| | - Shanice Lewis
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom
| | - Graham Pickard
- Department of Infection and Immunity Sciences Northwest London Pathology NHS Trust, Charing Cross Hospital, Fulham Palace Road W6 6RF, United Kingdom
| | - David Thomas
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, W12 0HS, United Kingdom
| | - Stephen P. McAdoo
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, W12 0HS, United Kingdom
| | - Liz Lightstone
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, W12 0HS, United Kingdom
| | - Alison Cox
- Department of Infection and Immunity Sciences Northwest London Pathology NHS Trust, Charing Cross Hospital, Fulham Palace Road W6 6RF, United Kingdom
| | - Peter Kelleher
- Department of Infection and Immunity Sciences Northwest London Pathology NHS Trust, Charing Cross Hospital, Fulham Palace Road W6 6RF, United Kingdom
- Department of Infectious Diseases, Imperial College London, Chelsea &Westminster Hospital Campus, Fulham Road London SW10 9NH, United Kingdom
| | - Michelle Willicombe
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, W12 0HS, United Kingdom
- Corresponding author at: Centre for Inflammatory Disease Department of Immunology and Inflammation, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, United Kingdom.
| |
Collapse
|
45
|
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: 60] [Impact Index Per Article: 20.0] [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
|
46
|
Cheng CH, Hung HC, Lee JC, Huang PW, Gu PW, Lai Y, Wang YC, Wu TH, Lee CF, Wu TJ, Chou HS, Chan KM, Huang CG, Lee WC. Determinants of Antibody Response to SARS-CoV-2 Vaccines in Liver Transplant Recipients: The Role of Immunosuppression Reduction. Vaccines (Basel) 2022; 10:1827. [PMID: 36366336 PMCID: PMC9692368 DOI: 10.3390/vaccines10111827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/23/2022] [Accepted: 10/25/2022] [Indexed: 11/29/2022] Open
Abstract
Liver transplant recipients on chronic immunosuppression show an attenuated antibody response after SARS-CoV-2 vaccination. Adjusting immunosuppressants during vaccination remains debated. We enrolled 380 liver transplant recipients receiving 2 doses of a protein subunit, mRNA, or a vector vaccine. The patients were informed to temporarily suspend immunosuppression for 2 weeks for both vaccination doses. We measured anti-live-SARS-CoV-2 spike neutralizing antibody levels at 1−2 months after the second vaccination; 83.9% of patients had humoral responses (SARS-CoV-2 NT50 ≥ 9.62 IU/mL) to 2 doses of vaccines. The mRNA (86.7%) and protein subunit vaccines (85%) yielded higher response rates than the vector vaccines (40.9%). Immunosuppression suspension during the two vaccinations yielded a higher response rate (91.5% vs. 57.7%). Only eight patients (2.1%) experienced transaminase level elevation of thrice the normal value (>110 IU/L) after the second vaccination. Most recovered spontaneously after resuming immunosuppression. Multivariate analysis revealed ABO incompatibility, white blood cell count <4000, lymphocyte count <20%, tacrolimus trough level >6.5 ng/mL, and no immunosuppression adjustment as independent risk factors to nonresponse. The mRNA and protein subunit vaccines yielded a higher response rate. Immunosuppression suspension for 2 weeks enhanced the antibody response. ABO incompatibility, leukopenia, lymphopenia, a high tacrolimus trough level, and no immunosuppression adjustment are associated with nonresponse.
Collapse
Affiliation(s)
- Chih-Hsien Cheng
- Department of General Surgery, Division of Liver and Transplantation Surgery, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Hao-Chien Hung
- Department of General Surgery, Division of Liver and Transplantation Surgery, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Jin-Chiao Lee
- Department of General Surgery, Division of Liver and Transplantation Surgery, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Po-Wei Huang
- Department of Laboratory Medicine, Chang-Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Po-Wen Gu
- Department of Laboratory Medicine, Chang-Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Yin Lai
- Department of General Surgery, Division of Liver and Transplantation Surgery, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Yu-Chao Wang
- Department of General Surgery, Division of Liver and Transplantation Surgery, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Tsung-Han Wu
- Department of General Surgery, Division of Liver and Transplantation Surgery, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Chen-Fang Lee
- Department of General Surgery, Division of Liver and Transplantation Surgery, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Ting-Jung Wu
- Department of General Surgery, Division of Liver and Transplantation Surgery, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Hong-Shiue Chou
- Department of General Surgery, Division of Liver and Transplantation Surgery, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Kun-Ming Chan
- Department of General Surgery, Division of Liver and Transplantation Surgery, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Chung-Guei Huang
- Department of Laboratory Medicine, Chang-Gung Memorial Hospital, Taoyuan 333, Taiwan
- Department of Medical Biotechnology and Laboratory Science, Chang-Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Wei-Chen Lee
- Department of General Surgery, Division of Liver and Transplantation Surgery, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| |
Collapse
|
47
|
Ponticelli C, Campise M. COVID-19 Vaccination in Kidney Transplant Candidates and Recipients. Vaccines (Basel) 2022; 10:vaccines10111808. [PMID: 36366317 PMCID: PMC9692413 DOI: 10.3390/vaccines10111808] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/14/2022] [Accepted: 10/20/2022] [Indexed: 12/03/2022] Open
Abstract
Kidney transplant candidates and kidney transplant recipients (KTRs) are at particular risk of severe complications of COVID-19 disease. In Western countries, mortality in affected hospitalized KTRs ranges between 19% and 50%. COVID-19 vaccination remains the most important measure to prevent the severity of infection in candidates and recipients of kidney transplant. However, the uraemic condition may affect the vaccine-induced immunity in patients with advanced chronic kidney disease (CKD) and in KTRs. Retention of uraemic toxins, dysbiosis, dysmetabolism, and dialysis can diminish the normal response to vaccination, leading to dysfunction of inflammatory and immune cells. In KTRs the efficacy of vaccines may be reduced by the immunosuppressive medications, and more than half of kidney transplant recipients are unable to build an immune response even after four administrations of anti-COVID-19 vaccines. The lack of antibody response leaves these patients at high risk for SARS-CoV-2 infection and severe COVID-19 disease. The aim of the present review is to focus on the main reasons for the impaired immunological response among candidates and kidney transplant recipients and to highlight some of the present options available to solve the problem.
Collapse
Affiliation(s)
| | - Mariarosaria Campise
- Department of Nephrology, Dialysis and Kidney Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Correspondence:
| |
Collapse
|
48
|
Predictors of Nonseroconversion to SARS-CoV-2 Vaccination in Kidney Transplant Recipients. Transplant Direct 2022; 8:e1397. [PMID: 36245996 PMCID: PMC9553374 DOI: 10.1097/txd.0000000000001397] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/04/2022] [Indexed: 11/27/2022] Open
Abstract
Kidney transplant recipients (KTRs) are still at risk of severe COVID-19 disease after SARS‑CoV‑2 vaccination, especially when they have limited antibody formation. Our aim was to understand the factors that may limit their humoral response.
Collapse
|
49
|
Osmanodja B, Stegbauer J, Kantauskaite M, Rump LC, Heinzel A, Reindl-Schwaighofer R, Oberbauer R, Benotmane I, Caillard S, Masset C, Kerleau C, Blancho G, Budde K, Grunow F, Mikhailov M, Schrezenmeier E, Ronicke S. Development and validation of multivariable prediction models of serological response to SARS-CoV-2 vaccination in kidney transplant recipients. Front Immunol 2022; 13:997343. [PMID: 36268021 PMCID: PMC9576943 DOI: 10.3389/fimmu.2022.997343] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/20/2022] [Indexed: 12/05/2022] Open
Abstract
Repeated vaccination against SARS-CoV-2 increases serological response in kidney transplant recipients (KTR) with high interindividual variability. No decision support tool exists to predict SARS-CoV-2 vaccination response to third or fourth vaccination in KTR. We developed, internally and externally validated five different multivariable prediction models of serological response after the third and fourth vaccine dose against SARS-CoV-2 in previously seronegative, COVID-19-naïve KTR. Using 20 candidate predictor variables, we applied statistical and machine learning approaches including logistic regression (LR), least absolute shrinkage and selection operator (LASSO)-regularized LR, random forest, and gradient boosted regression trees. For development and internal validation, data from 590 vaccinations were used. External validation was performed in four independent, international validation cohorts comprising 191, 184, 254, and 323 vaccinations, respectively. LASSO-regularized LR performed on the whole development dataset yielded a 20- and 10-variable model, respectively. External validation showed AUC-ROC of 0.840, 0.741, 0.816, and 0.783 for the sparser 10-variable model, yielding an overall performance 0.812. A 10-variable LASSO-regularized LR model predicts vaccination response in KTR with good overall accuracy. Implemented as an online tool, it can guide decisions whether to modulate immunosuppressive therapy before additional active vaccination, or to perform passive immunization to improve protection against COVID-19 in previously seronegative, COVID-19-naïve KTR.
Collapse
Affiliation(s)
- Bilgin Osmanodja
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Johannes Stegbauer
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Marta Kantauskaite
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Lars Christian Rump
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Andreas Heinzel
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University Vienna, Vienna, Austria
| | - Roman Reindl-Schwaighofer
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University Vienna, Vienna, Austria
| | - Rainer Oberbauer
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University Vienna, Vienna, Austria
| | - Ilies Benotmane
- Department of Nephrology and Transplantation, University Hospitals of Strasbourg, INSERM Unit 1109, Strasbourg, France
| | - Sophie Caillard
- Department of Nephrology and Transplantation, University Hospitals of Strasbourg, INSERM Unit 1109, Strasbourg, France
| | - Christophe Masset
- Institut de Transplantation Urologie Néphrologie, Centre Hospitalier Universitaire de Nantes, Centre de Recherche en Transplantation et Immunologie, UMR 1064, INSERM, Nantes Université, Nantes, France
| | - Clarisse Kerleau
- Institut de Transplantation Urologie Néphrologie, Centre Hospitalier Universitaire de Nantes, Centre de Recherche en Transplantation et Immunologie, UMR 1064, INSERM, Nantes Université, Nantes, France
| | - Gilles Blancho
- Institut de Transplantation Urologie Néphrologie, Centre Hospitalier Universitaire de Nantes, Centre de Recherche en Transplantation et Immunologie, UMR 1064, INSERM, Nantes Université, Nantes, France
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Fritz Grunow
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Michael Mikhailov
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Simon Ronicke
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
50
|
Impact of COVID-19 on the liver and on the care of patients with chronic liver disease, hepatobiliary cancer, and liver transplantation: An updated EASL position paper. J Hepatol 2022; 77:1161-1197. [PMID: 35868584 PMCID: PMC9296253 DOI: 10.1016/j.jhep.2022.07.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/06/2022] [Accepted: 07/06/2022] [Indexed: 02/06/2023]
Abstract
The COVID-19 pandemic has presented a serious challenge to the hepatology community, particularly healthcare professionals and patients. While the rapid development of safe and effective vaccines and treatments has improved the clinical landscape, the emergence of the omicron variant has presented new challenges. Thus, it is timely that the European Association for the Study of the Liver provides a summary of the latest data on the impact of COVID-19 on the liver and issues guidance on the care of patients with chronic liver disease, hepatobiliary cancer, and previous liver transplantation, as the world continues to deal with the consequences of the COVID-19 pandemic.
Collapse
|