1
|
Paranilam J, Arcioni F, Franco A, Lai KZH, Brown J, Kimball-Carroll S. Delphi Panel Consensus Statement Generation: COVID-19 Vaccination Recommendations for Immunocompromised Populations in the European Union. Infect Dis Ther 2024; 13:2227-2253. [PMID: 39382830 PMCID: PMC11499477 DOI: 10.1007/s40121-024-01051-9] [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: 06/17/2024] [Accepted: 09/09/2024] [Indexed: 10/10/2024] Open
Abstract
INTRODUCTION The coronavirus disease 2019 (COVID-19) pandemic has caused unprecedented pressure on healthcare systems globally. The lack of quality guidelines on the management of COVID-19 in rheumatologic disease, renal disease, hematological malignancy, and solid organ transplant recipients has resulted in a wide variation in clinical practice. METHODS Using a Delphi process, a panel of 16 key opinion leaders developed clinical practice statements regarding vaccine recommendations in areas where standards are absent or limited. Agreement among practicing physicians with consensus statements was also assessed via an online physician survey. The strength of the consensus was determined by the following rating system: a strong rating was defined as all four key opinion leaders (KOLs) rating the statement ≥ 8, a moderate rating was defined as three out of four KOLs rating the statement ≥ 8, and no consensus was defined as less than three out of four KOLs provided a rating of ≤ 8. Specialists voted on agreement with each consensus statement for their disease area using the same ten-point scoring system. RESULTS Key opinion leaders in rheumatology, nephrology, and hematology achieved consensuses for all nine statements pertaining to the primary and booster series with transplant physicians reaching consensus on eight of nine statements. Experts agreed that COVID-19 vaccines are safe, effective, and well tolerated by patients with rheumatological conditions, renal disease, hematologic malignancy, and recipients of solid organ transplants. The Delphi process yielded strong to moderate suggestions for the use of COVID-19 messenger ribonucleic acid (mRNA) vaccines and the necessity of the COVID-19 booster for the immunocompromised population. The expert panel had mixed feelings concerning the measurement of antibody titers, higher-dose mRNA vaccines, and the development of disease-specific COVID-19 guidance. CONCLUSIONS These results confirmed the necessity of COVID-19 vaccines and boosters in immunocompromised patients with rheumatologic disease, renal disease, hematological malignancy, and solid organ transplant recipients. Statements where consensus was not achieved were due to absent or limited evidence.
Collapse
Affiliation(s)
| | - Francesco Arcioni
- Pediatric Onco-Hematology with Bone Marrow Transplantation, Azienda Ospedaliera Di Perugia, Piazza Menghini 1, 06132, Perugia, Italy
| | - Antonio Franco
- Department of Nephrology, Hospital Dr Balmis, 03010, Alicante, Spain
| | | | | | | |
Collapse
|
2
|
Fujieda K, Tanaka A, Kikuchi R, Takai N, Saito S, Yasuda Y, Sano Y, Kato M, Furuhashi K, Maruyama S. Changes in antibody titer after four and five doses of the SARS-CoV-2 vaccine in Japanese post-kidney transplant patients. Ther Apher Dial 2024; 28:489-498. [PMID: 38385762 DOI: 10.1111/1744-9987.14114] [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: 10/29/2023] [Revised: 01/19/2024] [Accepted: 02/06/2024] [Indexed: 02/23/2024]
Abstract
INTRODUCTION Immunosuppressed patients exhibit low antibody acquisition rates following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination. Kidney transplant recipients previously exhibited low antibody acquisition rates after two vaccine doses, which increased after the third dose. We evaluated antibody titers of Japanese post-kidney transplant patients after the fourth and fifth vaccinations. METHODS Antibody titers for SARS-CoV-2 spike protein were measured between 3 weeks and 3 months after the fourth or fifth vaccination. RESULTS Increased antibody acquisition rates were observed after the fourth (75.0% antibody-positive) and fifth (81.5% antibody-positive) vaccinations. The antibody-acquired group after the fourth vaccination exhibited a higher body mass index and estimated glomerular filtration rate (eGFR) than the non-acquired group. A higher eGFR was associated with antibody acquisition after the fifth vaccination. CONCLUSION In Japanese post-kidney transplant patients, the antibody acquisition rate increased with each vaccine additional dose. Additional vaccinations are recommended to protect against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Kumiko Fujieda
- Department of Nephrology, Nagoya University Hospital, Nagoya, Japan
| | - Akihito Tanaka
- Department of Nephrology, Nagoya University Hospital, Nagoya, Japan
| | - Ryosuke Kikuchi
- Department of Medical Technique, Nagoya University Hospital, Nagoya, Japan
- Division of Clinical Laboratory, Gifu University Hospital, Gifu, Japan
| | - Nami Takai
- Department of Nursing, Nagoya University Hospital, Nagoya, Japan
| | - Shoji Saito
- Department of Nephrology, Nagoya University Hospital, Nagoya, Japan
| | - Yoshinari Yasuda
- Department of Nephrology, Nagoya University Hospital, Nagoya, Japan
| | - Yuta Sano
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masashi Kato
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Shoichi Maruyama
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
3
|
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; 108:00007890-990000000-00797. [PMID: 38902860 PMCID: PMC11581438 DOI: 10.1097/tp.0000000000005119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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
|
4
|
Bouwmans P, Malahe SRK, Messchendorp AL, Vart P, Imhof C, Sanders JSF, Gansevoort RT, de Vries APJ, Abrahams AC, Bemelman FJ, Vervoort JPM, Hilbrands LB, Ten Dam MAGJ, van den Dorpel RMA, Rispens T, Steenhuis M, Reinders MEJ, Hemmelder MH. Post COVID-19 condition imposes significant burden in patients with advanced chronic kidney disease: A nested case-control study. Int J Infect Dis 2024; 142:106990. [PMID: 38428480 DOI: 10.1016/j.ijid.2024.106990] [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: 11/21/2023] [Revised: 02/02/2024] [Accepted: 02/26/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND The burden of post COVID-19 condition (PCC) is not well studied in patients with advanced kidney disease. METHODS A large prospective cohort of SARS-CoV-2 vaccinated patients with chronic kidney disease stages G4-G5 (CKD G4/5), on dialysis, and kidney transplant recipients (KTR) were included. Antibody levels were determined after vaccination. Presence of long-lasting symptoms was assessed in patients with and without prior COVID-19 and compared using logistic regression. In patients with prior COVID-19, PCC was defined according to the WHO definition. RESULTS Two hundred sixteen CKD G4/5 patients, 375 dialysis patients, and 2005 KTR were included. Long-lasting symptoms were reported in 204/853 (24%) patients with prior COVID-19 and in 297/1743 (17%) patients without prior COVID-19 (aOR: 1.45 (1.17-1.78)], P < 0.001). PCC was prevalent in 29% of CKD G4/5 patients, 21% of dialysis patients, and 24% of KTR. In addition, 69% of patients with PCC reported (very) high symptom burden. Odds of PCC was lower per 10-fold increase in antibody level after vaccination (aOR 0.82 [0.70-0.96], P = 0.01) and higher in case of COVID-19 related hospital admission (aOR 4.64 [2.61-8.25], P = 0.003). CONCLUSIONS CKD G4/5 patients, dialysis patients, and KTR are at risk for PCC with high symptom burden after SARS-CoV-2 vaccination, especially if antibody levels are low and in case of hospitalization due to COVID-19.
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
| | - S Reshwan K Malahe
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - A Lianne Messchendorp
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, 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
| | - Céline Imhof
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan-Stephan F Sanders
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ron T Gansevoort
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, 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
| | - Alferso C Abrahams
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frederike J Bemelman
- Amsterdam UMC Location University of Amsterdam, Renal Transplant Unit, Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands
| | - Johanna P M Vervoort
- Dutch Kidney Patients Association (NVN), Bussum, The Netherlands; Department of Health Sciences, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Luuk B Hilbrands
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marc A G J Ten Dam
- Department of Internal Medicine, Canisius Wilhelmina Hospital, Nijmegen, 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
| | - Marlies E J Reinders
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Marc H Hemmelder
- 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
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Kitchens WH, Larsen CP, Badell IR. Costimulatory Blockade and Solid Organ Transplantation: The Past, Present, and Future. Kidney Int Rep 2023; 8:2529-2545. [PMID: 38106575 PMCID: PMC10719580 DOI: 10.1016/j.ekir.2023.08.037] [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: 04/03/2023] [Revised: 08/01/2023] [Accepted: 08/28/2023] [Indexed: 12/19/2023] Open
Abstract
Belatacept is the first costimulatory blockade agent clinically approved for transplant immunosuppression. Although more than 10 years of study have demonstrated that belatacept offers superior long-term renal allograft and patient survival compared to conventional calcineurin inhibitor (CNI)-based immunosuppression regimens, the clinical adoption of belatacept has continued to lag because of concerns of an early risk of acute cellular rejection (ACR) and various logistical barriers to its administration. In this review, the history of the clinical development of belatacept is examined, along with the findings of the seminal BENEFIT and BENEFIT-EXT trials culminating in the clinical approval of belatacept. Recent efforts to incorporate belatacept into novel CNI-free immunosuppression regimens are reviewed, as well as the experience of the Emory Transplant Center in using a tapered course of low-dose tacrolimus in belatacept-treated renal allograft patients to garner the long-term outcome benefits of belatacept without the short-term increased risks of ACR. Potential avenues to increase the clinical adoption of belatacept in the future are explored, including surmounting the logistical barriers of belatacept administration through subcutaneous administration or more infrequent belatacept dosing. In addition, belatacept conversion strategies and potential expanded clinical indications of belatacept are discussed for pediatric transplant recipients, extrarenal transplant recipients, treatment of antibody-mediated rejection (AMR), and in patients with failed renal allografts. Finally, we discuss the novel immunosuppressive drugs currently in the development pipeline that may aid in the expansion of costimulation blockade utilization.
Collapse
Affiliation(s)
- William H. Kitchens
- Division of Transplantation, Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Christian P. Larsen
- Division of Transplantation, Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - I. Raul Badell
- Division of Transplantation, Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
8
|
Christophorou E, Nilsson AC, Petersen I, Lindvig SO, Davidsen JR, Abazi R, Poulsen MK, Pedersen RM, Justesen US, Johansen NE, Bistrup C, Madsen LW, Johansen IS. Humoral antibody response following mRNA vaccines against SARS-CoV-2 in solid organ transplant recipients; a status after a fifth and bivalent vaccine dose. Front Immunol 2023; 14:1270814. [PMID: 38090591 PMCID: PMC10711048 DOI: 10.3389/fimmu.2023.1270814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/09/2023] [Indexed: 12/18/2023] Open
Abstract
Background In solid organ transplant (SOT) recipients, the humoral response following COVID-19 vaccination is reduced, as a result of their immunosuppressed treatment. In this study, we investigated antibody concentrations after booster vaccinations until the fifth dose, the latter by monovalent or bivalent BA1 or BA4/5 vaccines. In addition, we evaluated the efficacy of vaccination by recording breakthrough infections, hospitalizations, and deaths. Method This prospective cohort study included 438 SOT recipients (>18 years) vaccinated with mRNA vaccines against COVID-19 from January 2021 until March 2023. Blood samples were drawn before and after each vaccination and tested for SARS-CoV-2 spike RBD IgG antibodies with the lowest and highest cut-off at 7.1 and 5,680 BAU/mL, respectively. Vaccine information, breakthrough infections, and hospitalizations were collected from the medical records. Results Most participants received BNT162b2 and 61.4% received five vaccine doses. The response proportion in SOT recipients increased from 86.7% after the fourth dose to 93.0% following the fifth dose. Antibody concentration decreased with 142.7 BAU/mL between the third and fourth dose (median 132 days, Quartile 1: 123, Quartile 3: 148) and 234.3 BAU/mL between the fourth and fifth (median 250 days, Quartile 1: 241, Quartile 3: 262) dose among those without breakthrough infection (p=0.34). When comparing the Omicron BA.1 or Omicron BA.4/BA.5 adapted vaccines, no significant differences in antibody concentration were found, but 20.0% of SOT recipients receiving a monovalent fifth vaccine dose had a breakthrough infection compared to 4.0% and 7.9% among those who received BA.1 and BA.4/BA.5 adapted vaccines, respectively (p=0.04). Since January 2021, 240 (54.8%) participants had a breakthrough infection, and 22 were hospitalized, but no deaths were observed. Conclusions The fifth COVID-19 vaccine dose raised antibody response to 93.0% of the study population. Additional booster doses, as well as bivalent vaccines, led to higher levels of antibody concentration in SOT recipients. We found a lower incidence of breakthrough infections among SOT recipients after receiving a bivalent vaccine as a fifth dose compared to those receiving a monovalent dose. Antibody concentrations did not wane when the time between doses was prolonged from four to eight months.
Collapse
Affiliation(s)
- Emma Christophorou
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark
| | - Anna Christine Nilsson
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
| | - Inge Petersen
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark
| | - Susan O. Lindvig
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Jesper R. Davidsen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- South Danish Center for Interstitial Lung Diseases (SCILS), Department of Respiratory Medicine, Odense University Hospital, Odense, Denmark
| | - Rozeta Abazi
- Department of Gastroenterology, Odense University Hospital, Odense, Denmark
| | - Mikael K. Poulsen
- Department of Cardiology, Odense University Hospital, Odense, Denmark
| | - Rune M. Pedersen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Clinical Microbiology, Odense University Hospital, Odense, Denmark
| | - Ulrik S. Justesen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Clinical Microbiology, Odense University Hospital, Odense, Denmark
| | - Nicolai E. Johansen
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark
| | - Claus Bistrup
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Nephrology, Odense University Hospital, Odense, Denmark
| | - Lone W. Madsen
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
- Unit for Infectious Diseases, Department of Medicine, Lillebaelt Hospital, Kolding, Denmark
| | - Isik S. Johansen
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Open Patient Data Explorative Network, Odense University Hospital, Odense, Denmark
| |
Collapse
|
9
|
Stella C, Berardi C, Chiarito A, Gennenzi V, Postorino S, Settanni D, Cesarano M, Xhemalaj R, Tanzarella ES, Cutuli SL, Grieco DL, Conti G, Antonelli M, De Pascale G. Clinical features and 28-day mortality predictors of vaccinated patients admitted to a COVID-19 ICU hub in Italy. JOURNAL OF ANESTHESIA, ANALGESIA AND CRITICAL CARE 2023; 3:47. [PMID: 37957713 PMCID: PMC10644635 DOI: 10.1186/s44158-023-00130-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND COVID-19 vaccination has been proved to be effective in preventing hospitalization and illness progression, even though data on mortality of vaccinated patients in the intensive care unit (ICU) are conflicting. The aim of this study was to investigate the characteristics of vaccinated patients admitted to ICU according to their immunization cycle and to outline the risk factors for 28-day mortality. This observational study included adult patients admitted to ICU for acute respiratory failure (ARF) due to SARS-CoV-2 and who had received at least one dose of vaccine. RESULTS Fully vaccination was defined as a complete primary cycle from < 120 days or a booster dose from > 14 days. All the other patients were named partially vaccinated. One-hundred sixty patients (91 fully and 69 partially vaccinated) resulted eligible, showing a 28-day mortality rate of 51.9%. Compared to partially vaccinated, fully vaccinated were younger (69 [60-77.5] vs. 74 [66-79] years, p 0.029), more frequently immunocompromised (39.56% vs. 14.39%, p 0.003), and affected by at least one comorbidity (90.11% vs 78.26%, p 0.045), mainly chronic kidney disease (CKD) (36.26% vs 20.29%, p 0.035). At multivariable analysis, independent predictors of 28-day mortality were as follows: older age [OR 1.05 (CI 95% 1.01-1.08), p 0.005], history of chronic obstructive pulmonary disease (COPD) [OR 3.05 (CI 95% 1.28-7.30), p 0.012], immunosuppression [OR 3.70 (CI 95% 1.63-8.40), p 0.002], and admission respiratory and hemodynamic status [PaO2/FiO2 and septic shock: OR 0.99 (CI 95% 0.98-0.99), p 0.009 and 2.74 (CI 95% 1.16-6.48), p 0.022, respectively]. CONCLUSIONS Despite a full vaccination cycle, severe COVID-19 may occur in patients with relevant comorbidities, especially immunosuppression and CKD. Regardless the immunization status, predisposing conditions (i.e., older age, COPD, and immunosuppression) and a severe clinical presentation were predictors of 28-day mortality.
Collapse
Affiliation(s)
- Claudia Stella
- Dipartimento Di Scienze Biotecnologiche Di Base, Cliniche Intensivologiche E Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Cecilia Berardi
- Dipartimento Di Scienze Biotecnologiche Di Base, Cliniche Intensivologiche E Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Annalisa Chiarito
- Dipartimento Di Scienze Biotecnologiche Di Base, Cliniche Intensivologiche E Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Veronica Gennenzi
- Dipartimento Di Scienze Dell'Emergenza, Anestesiologiche E Della Rianimazione, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Stefania Postorino
- Dipartimento Di Scienze Dell'Emergenza, Anestesiologiche E Della Rianimazione, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Donatella Settanni
- Dipartimento Di Scienze Dell'Emergenza, Anestesiologiche E Della Rianimazione, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Melania Cesarano
- Dipartimento Di Scienze Dell'Emergenza, Anestesiologiche E Della Rianimazione, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Rikardo Xhemalaj
- Dipartimento Di Scienze Biotecnologiche Di Base, Cliniche Intensivologiche E Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Eloisa Sofia Tanzarella
- Dipartimento Di Scienze Dell'Emergenza, Anestesiologiche E Della Rianimazione, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Salvatore Lucio Cutuli
- Dipartimento Di Scienze Dell'Emergenza, Anestesiologiche E Della Rianimazione, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Domenico Luca Grieco
- Dipartimento Di Scienze Dell'Emergenza, Anestesiologiche E Della Rianimazione, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giorgio Conti
- Dipartimento Di Scienze Biotecnologiche Di Base, Cliniche Intensivologiche E Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento Di Scienze Dell'Emergenza, Anestesiologiche E Della Rianimazione, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Massimo Antonelli
- Dipartimento Di Scienze Biotecnologiche Di Base, Cliniche Intensivologiche E Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento Di Scienze Dell'Emergenza, Anestesiologiche E Della Rianimazione, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Gennaro De Pascale
- Dipartimento Di Scienze Biotecnologiche Di Base, Cliniche Intensivologiche E Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento Di Scienze Dell'Emergenza, Anestesiologiche E Della Rianimazione, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
10
|
Messchendorp AL, Gansevoort RT. Vaccinated kidney transplant recipients are yet not sufficiently protected against COVID-19. Nephrol Dial Transplant 2023; 38:2423-2425. [PMID: 37370226 PMCID: PMC10615622 DOI: 10.1093/ndt/gfad127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Indexed: 06/29/2023] Open
Affiliation(s)
- A Lianne Messchendorp
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ron T Gansevoort
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
11
|
Wang D, Zhou C, Wang C, Guo S, Zhang Y, Lv H, Zhou FH. COVID-19 Vaccine in Renal Transplant Recipients: A Bibliometric-Based Analysis of Trends. Transplant Proc 2023; 55:1771-1783. [PMID: 37481393 DOI: 10.1016/j.transproceed.2023.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/23/2023] [Indexed: 07/24/2023]
Abstract
BACKGROUND The global community has been affected by COVID-19, which emerged in December 2019. Since then, many studies have been conducted on kidney transplant recipients (KTRs) and COVID-19. This study aimed to perform a bibliometric and visual analysis of the published relationship between KTRs and COVID-19. OBJECTIVE To discuss the current status, hot spots, and development trend of research on KTRs vaccination with the COVID-19 vaccine and to provide a reference for researchers in related fields. METHODS Visual analysis of countries/regions, institutions, authors, references cited, and keywords for 2020 to 2023 via Microsoft Office Excel 2019 and CiteSpace (6.1.R6) based on the Web of Science core database. RESULTS A total of 366 publications were included after screening, with a rapid increase in the global literature studying the COVID-19 vaccine of KTRs. The US has the highest number of publications, indicating that it is the leading country in this field of research. Charite University of Medicine Berlin and Schrezenmeier E are the most published institutions and authors, respectively. "Antibody Response After a Third Dose of the messenger RNA-1273 SARS-CoV-2 Vaccine in Kidney Transplant Recipients With Minimal Serologic Response to 2 Doses" is the most central co-cited reference; The keywords "kidney transplant recipient," "covid 19 vaccine," and "mortality" have become hot topics of research. The keywords "humoral response" and "bnt162b2" are the latest research frontiers for detecting bursts. CONCLUSIONS This paper analyzed the current status and trends of vaccination studies in KTRs through bibliometric analysis. Several studies support the vaccination of KTRs with the COVID-19 vaccine. However, the evidence for improving vaccine efficacy by adjustment of immunosuppression is still limited, and future studies on vaccination will remain a hot topic in this field.
Collapse
Affiliation(s)
- Dong Wang
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, China; Department of Urology, Gansu Provincial Hospital, Lanzhou, China
| | - Chuan Zhou
- The First Clinical Medical College of Lanzhou University, Lanzhou, China; Department of Urology, Gansu Provincial Hospital, Lanzhou, China
| | - Chao Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, China; Department of Urology, Gansu Provincial Hospital, Lanzhou, China
| | - Sheng Guo
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, China; Department of Urology, Gansu Provincial Hospital, Lanzhou, China
| | - Yunfeng Zhang
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, China; Department of Urology, Gansu Provincial Hospital, Lanzhou, China
| | - Haoxuan Lv
- The First Clinical Medical College of Lanzhou University, Lanzhou, China; Department of Urology, Gansu Provincial Hospital, Lanzhou, China
| | - Feng-Hai Zhou
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, China; The First Clinical Medical College of Lanzhou University, Lanzhou, China; Department of Urology, Gansu Provincial Hospital, Lanzhou, China.
| |
Collapse
|
12
|
Hamaya T, Hatakeyama S, Yoneyama T, Tobisawa Y, Kodama H, Fujita T, Murakami R, Mori K, Okamoto T, Yamamoto H, Yoneyama T, Hashimoto Y, Saitoh H, Narumi S, Tomita H, Ohyama C. Humoral response to SARS-CoV-2 mRNA vaccine on in ABO blood type incompatible kidney transplant recipients treated with low-dose rituximab. Sci Rep 2023; 13:15098. [PMID: 37699969 PMCID: PMC10497504 DOI: 10.1038/s41598-023-42406-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/10/2023] [Indexed: 09/14/2023] Open
Abstract
We aimed to evaluate the humoral response after the second and third doses of SARS-CoV-2 mRNA vaccine in ABO blood type incompatible kidney transplant (KT) recipients treated with rituximab. This retrospective study conducted between June 2021 and June 2022 included 131 KT recipients and 154 nontransplant controls who had received mRNA vaccines. We compared the seropositivity (anti-SARS-CoV-2 spike IgG antibody titer ≥ 0.8 U/mL) after the second and third vaccinations. Furthermore, we evaluated the impact of pretransplant vaccination for seropositivity. Of the 131 KT recipients, 50 had received the third dose of mRNA vaccine. The antibody titer was significantly increased after the third dose of mRNA vaccine. The seropositivity rate after the third dose of mRNA vaccine increased from 36 to 70%. We observed no significant difference in seropositivity after the third dose of mRNA vaccine in ABO incompatibility, rituximab use, mycophenolate mofetil use, and age at KT. Of the nine recipients who had received the second or third dose of the mRNA vaccine prior to the KT, eight of the recipients were seropositive both before and after the KT. Our results suggest that ABO incompatibility or rituximab use was not significantly associated with seropositivity.
Collapse
Affiliation(s)
- Tomoko Hamaya
- Department of Urology, Hirosaki University School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Shingo Hatakeyama
- Department of Advanced Blood Purification Therapy, Hirosaki University School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan.
| | - Tohru Yoneyama
- Department of Glycotechnology, Center for Advanced Medical Research, Hirosaki University School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Yuki Tobisawa
- Department of Urology, Hirosaki University School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Hirotake Kodama
- Department of Urology, Hirosaki University School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Takeshi Fujita
- Department of Cardiology and Nephrology, Hirosaki University School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Reiichi Murakami
- Department of Cardiology and Nephrology, Hirosaki University School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Kazuyuki Mori
- Department of Urology, Hirosaki University School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Teppei Okamoto
- Department of Urology, Hirosaki University School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Hayato Yamamoto
- Department of Urology, Hirosaki University School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Takahiro Yoneyama
- Department of Advanced Transplant and Regenerative Medicine, Hirosaki University School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Yasuhiro Hashimoto
- Department of Urology, Hirosaki University School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Hisao Saitoh
- Department of Urology, Oyokyo Kidney Research Institute, 90 Kozawayamazaki, Hirosaki, Aomori, 036-8243, Japan
| | - Shunji Narumi
- Department of Transplant Nephrology and Surgery, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Nagoya, Japan
| | - Hirofumi Tomita
- Department of Cardiology and Nephrology, Hirosaki University School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Chikara Ohyama
- Department of Urology, Hirosaki University School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
- Department of Advanced Blood Purification Therapy, Hirosaki University School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
- Department of Advanced Transplant and Regenerative Medicine, Hirosaki University School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| |
Collapse
|
13
|
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
|
14
|
Frölke SC, Bouwmans P, Messchendorp AL, Vervoort JP, Abrahams AC, de Vries AP, Nieuwkerk PT, Hemmelder MH, Gansevoort RT, Hilbrands LB, Reinders ME, Sanders JSF, Bemelman FJ, Geerlings SE. Adherence to preventive measures after SARS-CoV-2 vaccination and after awareness of antibody response in kidney transplant recipients in the Netherlands: a nationwide questionnaire study. EClinicalMedicine 2023; 62:102103. [PMID: 37533418 PMCID: PMC10393559 DOI: 10.1016/j.eclinm.2023.102103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 06/21/2023] [Accepted: 06/29/2023] [Indexed: 08/04/2023] Open
Abstract
Background Kidney transplant recipients (KTRs) were advised to tightly adhere to government recommendations to curb the spread of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) because of a high risk of morbidity and mortality and decreased immunogenicity after vaccination. The aim of this study was to analyse the change in adherence to preventive measures after vaccination and awareness of antibody response, and to evaluate its effectiveness. Methods In this large-scale, national questionnaire study, questionnaires were sent to 3531 KTRs enrolled in the Dutch RECOVAC studies, retrospectively asking for adherence to nine preventive measures on a 5-point Likert scale before and after SARS-CoV-2 vaccination and after awareness of antibody response. Blood samples were collected 28 days after the second vaccination. Antibody response was categorised as non-responder (≤50 BAU/mL), low-responder (>50 ≤ 300 BAU/mL) or high-responder (>300 BAU/mL), and shared with participants as a correlate of protection. Participants of whom demographics on sex and age, blood samples and completed questionnaires were available, were included. Our study took place between February 2021 and January 2022. The primary outcome of adherence before and after vaccination was assessed between August and October 2021 and compared via the Wilcoxon signed rank sum test. Logistic regression analysis was performed to estimate the association between antibody response and non-adherence, and adherence on acquiring SARS-CoV-2 infection. This study is registered at ClinicalTrials.gov (NCT04841785). Findings In 2939 KTRs (83%) who completed the first questionnaire on adherence to preventive measures, adherence was higher before than after vaccination (4.56, IQR 4.11-4.78 and 4.22, IQR 3.67-4.67, p < 0.001). Adherence after awareness of antibody response was analysed in 2399 KTRs (82%) of whom also blood samples were available, containing 949 non-responders, 500 low-responders and 950 high-responders. Compared to non-responders, low- and high-responders reported higher non-adherence. Higher adherence was associated with lower infection rates before and after vaccination (OR 0.67 [0.51-0.91], p = 0.008 and OR 0.48 [0.28-0.86], p = 0.010). Interpretation Adherence decreased after SARS-CoV-2 vaccination and in KTRs who were aware of a subsequent antibody response compared with those without. Preventive measures in this vulnerable group seem to be effective, regardless of vaccination status. This study starts a debate on sharing antibody results with the patient and future studies should elucidate whether decreased adherence in antibody responders is justified, also in view of future pandemics. Funding The Netherlands Organization for Health Research and Development and the Dutch Kidney Foundation.
Collapse
Affiliation(s)
- Sophie C. Frölke
- 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
| | - Pim Bouwmans
- Division of Nephrology, Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - A. Lianne Messchendorp
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Johanna P.M. Vervoort
- Dutch Kidney Patient Association (NVN), Bussum, the Netherlands
- Global Health Unit, Department of Health Sciences, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Alferso C. Abrahams
- Department of Nephrology and Hypertension, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Aiko P.J. de Vries
- Department of Medicine, Division of Nephrology, Leiden University Medical Centre, Leiden, the Netherlands
- Leiden Transplant Centre, Leiden, the Netherlands
| | - Pythia T. Nieuwkerk
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
- Amsterdam UMC Location University of Amsterdam, Medical Psychology, Meibergdreef 9, Amsterdam, the Netherlands
- Amsterdam Public Health, Quality of Care, Amsterdam, the Netherlands
| | - Marc H. Hemmelder
- Division of Nephrology, Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Ron T. Gansevoort
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Luuk B. Hilbrands
- Department of Nephrology, Radboud University Medical Centre, Radboud Institute for Health Sciences, Nijmegen, the Netherlands
| | - Marlies E.J. Reinders
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Jan-Stephan F. Sanders
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Centre 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
| | - Suzanne E. Geerlings
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
- Amsterdam Public Health, Quality of Care, Amsterdam, the Netherlands
- Amsterdam UMC Location University of Amsterdam, Infectious Diseases, Meibergdreef 9, Amsterdam, the Netherlands
| | | |
Collapse
|
15
|
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
|
16
|
Housset P, Bouhelier C, Pardon A, Hanafi L, Vittoz N, Bozman DF, Amar W, Caudwell V, Charlemagne T. Bivalent vaccination against COVID-19 can trigger a humoral response in immunocompromised patients who receive monoclonal antibody prophylaxis. J Nephrol 2023:10.1007/s40620-023-01699-5. [PMID: 37410357 DOI: 10.1007/s40620-023-01699-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/09/2023] [Indexed: 07/07/2023]
Affiliation(s)
- Pierre Housset
- Department of Nephrology, Centre Hospitalier Sud-Francilien, 40 Avenue Serge Dassault, 91100, Corbeil-Essonnes, France.
| | - Christine Bouhelier
- Department of Nephrology, Centre Hospitalier Sud-Francilien, 40 Avenue Serge Dassault, 91100, Corbeil-Essonnes, France
| | - Agathe Pardon
- Department of Nephrology, Centre Hospitalier Sud-Francilien, 40 Avenue Serge Dassault, 91100, Corbeil-Essonnes, France
| | - Latifa Hanafi
- Department of Nephrology, Centre Hospitalier Sud-Francilien, 40 Avenue Serge Dassault, 91100, Corbeil-Essonnes, France
| | - Nathalie Vittoz
- Department of Nephrology, Centre Hospitalier Sud-Francilien, 40 Avenue Serge Dassault, 91100, Corbeil-Essonnes, France
| | - Dogan-Firat Bozman
- Department of Nephrology, Centre Hospitalier Sud-Francilien, 40 Avenue Serge Dassault, 91100, Corbeil-Essonnes, France
| | - Wassila Amar
- Department of Nephrology, Centre Hospitalier Sud-Francilien, 40 Avenue Serge Dassault, 91100, Corbeil-Essonnes, France
| | - Valérie Caudwell
- Department of Nephrology, Centre Hospitalier Sud-Francilien, 40 Avenue Serge Dassault, 91100, Corbeil-Essonnes, France
| | - Thibaut Charlemagne
- Department of Nephrology, Centre Hospitalier Sud-Francilien, 40 Avenue Serge Dassault, 91100, Corbeil-Essonnes, France
| |
Collapse
|
17
|
Cazorla JM, Villanego F, Aguilera A, Garcia T, Orellana C, Trujillo T, Gómez AM, Mazuecos A. Humoral Response After Five Successive Doses of SARS-CoV-2 mRNA Vaccine in Kidney Transplant Patients. Transplantation 2023; 107:e188-e189. [PMID: 37170412 DOI: 10.1097/tp.0000000000004628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Affiliation(s)
- Juan Manuel Cazorla
- Department of Nephrology, Hospital Universitario Puerta del Mar, Cadiz, Spain
| | | | - Aurora Aguilera
- Department of Nephrology, Hospital Universitario Puerta del Mar, Cadiz, Spain
| | - Teresa Garcia
- Department of Nephrology, Hospital Universitario Puerta del Mar, Cadiz, Spain
| | - Cristhian Orellana
- Department of Nephrology, Hospital Universitario Puerta del Mar, Cadiz, Spain
| | - Teresa Trujillo
- Department of Microbiology, Hospital Universitario Puerta del Mar, Cadiz, Spain
| | - Ana María Gómez
- Department of Nephrology, Hospital Universitario Puerta del Mar, Cadiz, Spain
| | | |
Collapse
|
18
|
Meredith RT, Bermingham MD, Bentley K, Agah S, Aboagye-Odei A, Yarham RAR, Mills H, Shaikh M, Hoye N, Stanton RJ, Chadwick DR, Oliver MA. Differential cellular and humoral immune responses in immunocompromised individuals following multiple SARS-CoV-2 vaccinations. Front Cell Infect Microbiol 2023; 13:1207313. [PMID: 37424787 PMCID: PMC10327606 DOI: 10.3389/fcimb.2023.1207313] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction The heterogeneity of the immunocompromised population means some individuals may exhibit variable, weak or reduced vaccine-induced immune responses, leaving them poorly protected from COVID-19 disease despite receiving multiple SARS-CoV-2 vaccinations. There is conflicting data on the immunogenicity elicited by multiple vaccinations in immunocompromised groups. The aim of this study was to measure both humoral and cellular vaccine-induced immunity in several immunocompromised cohorts and to compare them to immunocompetent controls. Methods Cytokine release in peptide-stimulated whole blood, and neutralising antibody and baseline SARS-CoV-2 spike-specific IgG levels in plasma were measured in rheumatology patients (n=29), renal transplant recipients (n=46), people living with HIV (PLWH) (n=27) and immunocompetent participants (n=64) post third or fourth vaccination from just one blood sample. Cytokines were measured by ELISA and multiplex array. Neutralising antibody levels in plasma were determined by a 50% neutralising antibody titre assay and SARS-CoV-2 spike specific IgG levels were quantified by ELISA. Results In infection negative donors, IFN-γ, IL-2 and neutralising antibody levels were significantly reduced in rheumatology patients (p=0.0014, p=0.0415, p=0.0319, respectively) and renal transplant recipients (p<0.0001, p=0.0005, p<0.0001, respectively) compared to immunocompetent controls, with IgG antibody responses similarly affected. Conversely, cellular and humoral immune responses were not impaired in PLWH, or between individuals from all groups with previous SARS-CoV-2 infections. Discussion These results suggest that specific subgroups within immunocompromised cohorts could benefit from distinct, personalised immunisation or treatment strategies. Identification of vaccine non-responders could be critical to protect those most at risk.
Collapse
Affiliation(s)
| | | | - Kirsten Bentley
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Sayeh Agah
- InBio, Charlottesville, VA, United States
| | - Abigail Aboagye-Odei
- Department of Infectious Diseases, South Tees Hospitals National Health Service (NHS) Foundation Trust, Middlesbrough, England, United Kingdom
| | | | | | - Muddassir Shaikh
- Department of Kidney Services, South Tees Hospitals National Health Service (NHS) Foundation Trust, Middlesbrough, England, United Kingdom
| | - Neil Hoye
- Department of Rheumatology, South Tees Hospitals National Health Service (NHS) Foundation Trust, Middlesbrough, England, United Kingdom
| | - Richard J. Stanton
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - David R. Chadwick
- Department of Infectious Diseases, South Tees Hospitals National Health Service (NHS) Foundation Trust, Middlesbrough, England, United Kingdom
| | | |
Collapse
|
19
|
Li HJ, Yang QC, Yao YY, Huang CY, Yin FQ, Xian-Yu CY, Zhang C, Chen SJ. COVID-19 vaccination effectiveness and safety in vulnerable populations: a meta-analysis of 33 observational studies. Front Pharmacol 2023; 14:1144824. [PMID: 37426814 PMCID: PMC10326898 DOI: 10.3389/fphar.2023.1144824] [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: 01/15/2023] [Accepted: 06/15/2023] [Indexed: 07/11/2023] Open
Abstract
Background: Even 3 years into the COVID-19 pandemic, questions remain about how to safely and effectively vaccinate vulnerable populations. A systematic analysis of the safety and efficacy of the COVID-19 vaccine in at-risk groups has not been conducted to date. Methods: This study involved a comprehensive search of PubMed, EMBASE, and Cochrane Central Controlled Trial Registry data through 12 July 2022. Post-vaccination outcomes included the number of humoral and cellular immune responders in vulnerable and healthy populations, antibody levels in humoral immune responders, and adverse events. Results: A total of 23 articles assessing 32 studies, were included. The levels of IgG (SMD = -1.82, 95% CI [-2.28, -1.35]), IgA (SMD = -0.37, 95% CI [-0.70, -0.03]), IgM (SMD = -0.94, 95% CI [-1.38, -0.51]), neutralizing antibodies (SMD = -1.37, 95% CI [-2.62, -0.11]), and T cells (SMD = -1.98, 95% CI [-3.44, -0.53]) were significantly lower in vulnerable than in healthy populations. The positive detection rates of IgG (OR = 0.05, 95% CI [0.02, 0.14]) and IgA (OR = 0.03, 95% CI [0.01, 0.11]) antibodies and the cellular immune response rates (OR = 0.20, 95% CI [0.09, 0.45]) were also lower in the vulnerable populations. There were no statistically significant differences in fever (OR = 2.53, 95% CI [0.11, 60.86]), chills (OR = 2.03, 95% CI [0.08, 53.85]), myalgia (OR = 10.31, 95% CI [0.56, 191.08]), local pain at the injection site (OR = 17.83, 95% CI [0.32, 989.06]), headache (OR = 53.57, 95% CI [3.21, 892.79]), tenderness (OR = 2.68, 95% CI [0.49, 14.73]), and fatigue (OR = 22.89, 95% CI [0.45, 1164.22]) between the vulnerable and healthy populations. Conclusion: Seroconversion rates after COVID-19 vaccination were generally worse in the vulnerable than healthy populations, but there was no difference in adverse events. Patients with hematological cancers had the lowest IgG antibody levels of all the vulnerable populations, so closer attention to these patients is recommended. Subjects who received the combined vaccine had higher antibody levels than those who received the single vaccine.
Collapse
Affiliation(s)
- Hui-Jun Li
- Center for Evidence-Based Medicine and Clinical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Qi-Chao Yang
- Department of Emergency, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yang-Yang Yao
- Center for Evidence-Based Medicine and Clinical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Cheng-Yang Huang
- Center for Evidence-Based Medicine and Clinical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Fu-Qiang Yin
- Center for Evidence-Based Medicine and Clinical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Chen-Yang Xian-Yu
- Center for Evidence-Based Medicine and Clinical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Chao Zhang
- Center for Evidence-Based Medicine and Clinical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Shao-Juan Chen
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| |
Collapse
|
20
|
Kawabe M, Kuroda T, Yamamoto I, Kobayashi A, Ohki Y, Hayashi A, Urabe F, Miki J, Yamada H, Kimura T, Matsuo N, Tanno Y, Horino T, Ohkido I, Yamamoto H, Yokoo T. Booster effect of the third dose of SARS-CoV-2 mRNA vaccine in Japanese kidney transplant recipients. Sci Rep 2023; 13:9976. [PMID: 37340001 DOI: 10.1038/s41598-023-36998-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 06/14/2023] [Indexed: 06/22/2023] Open
Abstract
The humoral response of kidney transplant recipients (KTR) to the mRNA vaccine against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is generally poor. We evaluated the booster effect of the third dose (D3) of two SARS-CoV-2 mRNA vaccines 6 months after the second dose (D2) in Japanese KTR. The anti-spike (anti-S) antibody titer 1 and 3 months after the D3 was evaluated in 82 Japanese KTR. The primary endpoint was the seropositivity rate, and factors associated with the lack of a response were evaluated in a logistic regression model. Overall, the anti-S antibody seropositivity rate 1 and 3 months after the D3 was 74.7% and 76.0%. The anti-S antibody titers after the first and second doses were higher in patients vaccinated with the mRNA-1273 than with the BNT162b2 vaccine. Among the 38 KTR who were seronegative 5 months after the D2, 18 (47.4%) became seropositive following the D3. Factors associated with a non-response were mycophenolic acid dose, post-transplant duration, hemoglobin, and lymphocyte count. A humoral response 1 and 3 months after the D3 was obtained in ~ 75% of KTR, but 20% were non-responders. Additional studies are needed to clarify the factors hindering a vaccine response.
Collapse
Affiliation(s)
- Mayuko Kawabe
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Takafumi Kuroda
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Izumi Yamamoto
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan.
| | - Akimitsu Kobayashi
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yutaro Ohki
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Ayaka Hayashi
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Fumihiko Urabe
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Jun Miki
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiroki Yamada
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Takahiro Kimura
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Nanae Matsuo
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yudo Tanno
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Tetsuya Horino
- Department of Infectious Disease and Infection Control, The Jikei University School of Medicine, Tokyo, Japan
| | - Ichiro Ohkido
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiroyasu Yamamoto
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Takashi Yokoo
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
21
|
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: 3] [Impact Index Per Article: 1.5] [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
|
22
|
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: 5] [Impact Index Per Article: 2.5] [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
|
23
|
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
|
24
|
Deguchi H, Sakamoto A, Nakamura N, Okabe Y, Miura Y, Iida T, Yoshimura M, Haga N, Nabeshima S, Masutani K. Antibody acquisition after second and third SARS-CoV-2 vaccinations in Japanese kidney transplant patients: a prospective study. Clin Exp Nephrol 2023; 27:574-582. [PMID: 36914824 PMCID: PMC10010649 DOI: 10.1007/s10157-023-02334-0] [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: 01/31/2023] [Accepted: 02/27/2023] [Indexed: 03/14/2023]
Abstract
BACKGROUND Kidney transplant patients have lower antibody acquisition after SARS-CoV-2 vaccination. The efficacy of vaccines in Japanese kidney transplant patients with specific characteristics, such as predominant living-donor, ABO-incompatible kidney transplant, and low-dose immunosuppression, requires verification. METHODS We conducted a prospective study to estimate anti-SARS-CoV-2 antibody levels in 105 kidney transplant patients and 57 controls. Blood samples were obtained before vaccination, 1, 3, and 6 months after second vaccination, and 1 month after third vaccination. We investigated antibody acquisition rates, antibody levels, and factors associated with antibody acquisition. RESULTS One month after second vaccination, antibody acquisition was 100% in the controls but only 36.7% in the kidney transplant group (P < 0.001). Antibody levels in positive kidney transplant patients were also lower than in the controls (median, 4.9 arbitrary units vs 106.4 arbitrary units, respectively, P < 0.001). Years after kidney transplant (odds ratio 1.107, 95% confidence interval 1.012-1.211), ABO-incompatible kidney transplant (odds ratio 0.316, 95% confidence interval 0.101-0.991) and mycophenolate mofetil use (odds ratio 0.177, 95% confidence interval 0.054-0.570) were significant predictors for antibody acquisition after second vaccination. After third vaccination, antibody positivity in the kidney transplant group increased to 75.3%, and antibody levels in positive patients were 71.7 arbitrary units. No factors were associated with de novo antibody acquisition. CONCLUSIONS In Japanese kidney transplant patients, years after kidney transplant, ABO-incompatible kidney transplant and mycophenolate mofetil use were predictors for antibody acquisition after second vaccination. Third vaccination improves antibody status even in patients who were seronegative after the second vaccination.
Collapse
Affiliation(s)
- Hidetaka Deguchi
- Division of Nephrology and Rheumatology, Department of Internal Medicine, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-Ku, Fukuoka, 814-0180, Japan
| | - Atsuhiko Sakamoto
- Department of General Medicine, Fukuoka University Hospital, Fukuoka, Japan
| | - Nobuyuki Nakamura
- Department of Urology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Yasuhiro Okabe
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshifumi Miura
- Department of General Surgery, Hara-Sanshin Hospital, Fukuoka, Japan
| | | | - Michinobu Yoshimura
- Department of Microbiology and Immunology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Nobuhiro Haga
- Department of Urology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Shigeki Nabeshima
- Department of General Medicine, Fukuoka University Hospital, Fukuoka, Japan
| | - Kosuke Masutani
- Division of Nephrology and Rheumatology, Department of Internal Medicine, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-Ku, Fukuoka, 814-0180, Japan.
| |
Collapse
|
25
|
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: 32] [Impact Index Per Article: 16.0] [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
|
26
|
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: 4] [Impact Index Per Article: 2.0] [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
|
27
|
Park JK, Bhandari S. Literature review on COVID-19 vaccine efficacy in the immunocompromised population, and possible implications to future vaccination in kidney transplant patients. Front Med (Lausanne) 2023; 10:1103699. [PMID: 36817787 PMCID: PMC9931721 DOI: 10.3389/fmed.2023.1103699] [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: 11/20/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Since the emergence of the virulent coronavirus in 2019, efforts to tackle the coronavirus-disease-2019 (COVID-19) pandemic have been made globally. The development of the coronavirus disease (COVID) vaccine was a significant breakthrough in ways to tackle the virus. Various research studies have been conducted to identify how the virus works and ways to manage COVID, including the efficacy of the vaccines. However, there is limited data on how these measures work for the immunocompromised, despite the grave impact of these virulent strains in this population. Specifically, this review aims to focus on kidney transplant recipients (KTRs). Studies have suggested that there is significantly lower vaccine response in some immunocompromised groups despite additional booster doses, and hence warrants an augmented or alternative protection against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) for these patients. This suggests a need for alternative or more tailored approach in providing adequate protection against the COVID-19 in these cohorts. Some suggested ways include withholding immunosuppressants before and/or after vaccination, increasing the vaccine doses or reducing intervals and providing a mixture of monoclonal antibody (mAb) or antiviral therapy. However, the appropriate degree of alteration and augmentation, as well as its safety and effectiveness remains to be determined. Furthermore, continuous emergence of more virulent strains, such as the Omicron and its sub-lineages or the Deltacron, emphasises the need for ongoing research to assess the effectiveness of the current treatment against these new variants. Overall, active interest and appropriate updates to COVID-19 guidelines is necessary.
Collapse
Affiliation(s)
- Joo Kyung Park
- Department of Renal Medicine, Hull University Teaching Hospitals NHS Trust, Kingston upon Hull, United Kingdom,Joo Kyung Park, ; orcid.org/0000-0002-7034-4998
| | - Sunil Bhandari
- Department of Renal Medicine, Hull University Teaching Hospitals NHS Trust, Kingston upon Hull, United Kingdom,Hull York Medical School, Kingston upon Hull, United Kingdom,*Correspondence: Sunil Bhandari, ; orcid.org/0000-0002-0996-9622
| |
Collapse
|
28
|
Yakubu I, Moinuddin I, Gupta G. Use of belatacept in kidney transplantation: what's new? Curr Opin Organ Transplant 2023; 28:36-45. [PMID: 36326538 DOI: 10.1097/mot.0000000000001033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
PURPOSE OF REVIEW The advent of calcineurin inhibitors have led to a significant improvement in short term outcomes after kidney transplantation. However, long term outcomes are hindered by the cardiovascular, metabolic and chronic renal toxicity associated with these agents. Belatacept is a selective T cell costimulation blocker that is approved for prevention of rejection in kidney transplantation, and has been associated with favorable cardiovascular, metabolic and renal outcomes in kidney transplant recipients. This review provides an overview of recent updates in the use of belatacept in kidney transplant recipients. RECENT FINDINGS Belatacept may be a safe alternative to calcineurin inhibitors for select kidney transplant populations. Patients converted to belatacept from a calcineurin inhibitor-based immunosuppression generally experience improvement in renal function, and may be less likely to develop de novo donor specific antibodies or new onset diabetes after transplantation. Although, belatacept based immunosuppression may increase the risk of early acute cellular rejection, it may however be beneficial in stabilization of long-term renal function and improvement in inflammation in patients with chronic active antibody mediated rejection. These benefits need to be counterweighed with risks of lack of response to severe acute respiratory syndrome coronavirus 2 vaccination and other adverse infectious outcomes. SUMMARY Belatacept may be an alternative to calcineurin inhibitors and may contribute to improved long term metabolic and allograft outcomes in kidney transplant recipients. Careful selection of patients for belatacept-based immunosuppression is needed, to obviate the risk of acute rejection shown in clinical studies.
Collapse
Affiliation(s)
| | - Irfan Moinuddin
- Division of Nephrology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Gaurav Gupta
- Division of Nephrology, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
29
|
Mahallawi WH, Alharbi WA, Aloufi SA, Ibrahim NA, Abdelrahman MM, Alhomayeed BA, Aboonq MS, Alqahtani SAM, Rajih ES, Bakhsh AM, Sandokji I. Declined Humoral Immunity of Kidney Transplant Recipients to SARS-CoV-2 Vaccines. Infect Drug Resist 2023; 16:2829-2840. [PMID: 37193301 PMCID: PMC10182766 DOI: 10.2147/idr.s408686] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 05/03/2023] [Indexed: 05/18/2023] Open
Abstract
Background Kidney transplant recipients (KTRs) commonly suffer from impaired immunity. KTRs' compromised immune response to COVID-19 vaccines indicates urgent revision of immunisation policies. Methods A cross-sectional study was conducted in Madinah, Saudi Arabia of 84 KTRs who had received at least one dose of a COVID-19 vaccine. ELISA was used to evaluate anti-spike SARS-CoV-2 IgG and IgM antibody levels in blood samples obtained one month and seven months after vaccination. Univariate and multivariate analyses were performed to identify associations between seropositive status and factors such as the number of vaccine doses, transplant age, and immunosuppressive therapies. Results The mean age of KTRs was 44.3 ± 14.7 years. The IgG antibody seropositivity rate (n=66, 78.5%) was significantly higher than the seronegativity rate (n=18, 21.4%) in the whole cohort (p<0.001). In KTRs seroconverting after one month (n=66), anti-SARS-CoV-2 IgG levels declined significantly between one month (median [IQR]:3 [3-3]) and seven months (2.4 [1.7-2.6]) after vaccination (p<0.01). In KTRs with hypertension, IgG levels significantly decreased between one and seven months after vaccination (p<0.01). IgG levels also decreased significantly in KTRs with a transplant of >10 years (p=0.02). Maintenance immunosuppressive regimens (triple immunosuppressive therapy and steroid-based and antimetabolite-based regimens) led to a significant decrease in IgG levels between the first and second sample (p<0.01). KTRs receiving three vaccine doses showed higher antibody levels than those receiving a single dose or two doses, but the levels decreased significantly between one (median [IQR]: 3 [3-3]) and seven months (2.4 [1.9-2.6]) after vaccination (p<0.01). Conclusion KTRs' humoral response after SARS-CoV-2 vaccination is dramatically inhibited and wanes. Antibody levels show a significant decline over time in KTRs with hypertension; receiving triple immunosuppressive therapy or steroid-based or antimetabolite-based regimens; receiving mixed mRNA and viral vector vaccines; and with a transplant of >10 years.
Collapse
Affiliation(s)
- Waleed H Mahallawi
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia
- Correspondence: Waleed H Mahallawi, Medical Laboratory Technology Department, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia, Email
| | - Wael A Alharbi
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia
| | - Sultan A Aloufi
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia
| | - Nadir A Ibrahim
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia
| | | | | | - Moutasem S Aboonq
- Department of Physiology, College of Medicine, Taibah University, Madinah, Saudi Arabia
| | | | - Emad S Rajih
- Urology Department, College of Medicine, Taibah University, Madinah, Saudi Arabia
| | - Abdulaziz M Bakhsh
- Urology Department, College of Medicine, Taibah University, Madinah, Saudi Arabia
| | - Ibrahim Sandokji
- Department of Pediatrics, College of Medicine, Taibah University, Madinah, Saudi Arabia
| |
Collapse
|
30
|
Kodali L, Budhiraja P, Gea-Banacloche J. COVID-19 in kidney transplantation-implications for immunosuppression and vaccination. Front Med (Lausanne) 2022; 9:1060265. [PMID: 36507509 PMCID: PMC9727141 DOI: 10.3389/fmed.2022.1060265] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2022] Open
Abstract
COVID-19 pandemic continues to challenge the transplant community, given increased morbidity and mortality associated with the disease and poor response to prevention measures such as vaccination. Transplant recipients have a diminished response to both mRNA and vector-based vaccines compared to dialysis and the general population. The currently available assays to measure response to vaccination includes commercially available antibody assays for anti-Spike Ab, or anti- Receptor Binding Domain Ab. Positive antibody testing on the assays does not always correlate with neutralizing antibodies unless the antibody levels are high. Vaccinations help with boosting polyfunctional CD4+ T cell response, which continues to improve with subsequent booster doses. Ongoing efforts to improve vaccine response by using additional booster doses and heterologous vaccine combinations are underway. There is improved antibody response in moderate responders; however, the ones with poor response to initial vaccination doses, continue to have a poor response to sequential boosters. Factors associated with poor vaccine response include diabetes, older age, specific immunosuppressants such as belatacept, and high dose mycophenolate. In poor responders, a decrease in immunosuppression can increase response to vaccination. COVID infection or vaccination has not been associated with an increased risk of rejection. Pre- and Post-exposure monoclonal antibodies are available to provide further protection against COVID infection, especially in poor vaccine responders. However, the efficacy is challenged by the emergence of new viral strains. A recently approved bivalent vaccine offers better protection against the Omicron variant.
Collapse
Affiliation(s)
- Lavanya Kodali
- Department of Internal Medicine, Mayo Clinic, Phoenix, AZ, United States
- Division of Nephrology, Transplant Center, Mayo Clinic, Phoenix, AZ, United States
| | - Pooja Budhiraja
- Department of Internal Medicine, Mayo Clinic, Phoenix, AZ, United States
- Division of Nephrology, Transplant Center, Mayo Clinic, Phoenix, AZ, United States
| | - Juan Gea-Banacloche
- Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| |
Collapse
|
31
|
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
|
32
|
Babel N, Hugo C, Westhoff TH. Vaccination in patients with kidney failure: lessons from COVID-19. Nat Rev Nephrol 2022; 18:708-723. [PMID: 35999285 PMCID: PMC9397175 DOI: 10.1038/s41581-022-00617-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [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
|
33
|
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
|
34
|
Comparison of the Immune Response After an Extended Primary Series of COVID-19 Vaccination in Kidney Transplant Recipients Receiving Standard Versus Mycophenolic Acid-sparing Immunosuppressive Regimen. Transplant Direct 2022; 8:e1393. [PMID: 36312516 PMCID: PMC9605792 DOI: 10.1097/txd.0000000000001393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/29/2022] Open
Abstract
UNLABELLED Two doses of coronavirus disease 2019 vaccination provide suboptimal immune response in transplant patients. Mycophenolic acid (MPA) is one of the most important factors that blunts the immune response. We studied the immune response to the extended primary series of 2 doses of AZD1222 and a single dose of BNT162b2 in kidney transplant patients who were on the standard immunosuppressive regimen compared to those on the MPA-sparing regimen. METHODS The kidney transplant recipients who were enrolled into the study were divided into 2 groups based on their immunosuppressive regimen. Those on the standard immunosuppressive regimen received tacrolimus (TAC), MPA, and prednisolone (standard group). The patients in the MPA-sparing group received mammalian target of rapamycin inhibitors (mTORi) with low dose TAC plus prednisolone (MPA-sparing group). The vaccination consisted of 2 doses of AZD1222 and a single dose of BNT162b2. RESULTS A total of 115 patients completed the study. There were 76 (66.08%) patients in the standard group and 39 (33.91%) patients in the MPA-sparing group. The overall median anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) S antibody level at 4 wk after vaccine completion was 676.64 (interquartile range = 6.02-3644.03) BAU/mL with an 80% seroconversion rate. The MPA-sparing group achieved higher anti-SARS-CoV-2 S antibody level compared to the standard group (3060.69 and 113.91 BAU/mL, P < 0.001). The seroconversion rate of MPA-sparing and standard groups were 97.4% and 71.1%, respectively (P < 0.001). The anti-HLA antibodies did not significantly increase after vaccination. CONCLUSIONS The extended primary series of 2 doses of AZD1222 and a single dose of BNT162b2 provided significant humoral immune response. The MPA-sparing regimen with mTORi and low dose TAC had a higher ant-SARS-CoV-2 S antibody level and seroconversion rate compared to the participants in the standard regimen.
Collapse
|
35
|
Banjongjit A, Phirom S, Phannajit J, Jantarabenjakul W, Paitoonpong L, Kittanamongkolchai W, Wattanatorn S, Prasithsirikul W, Eiam-Ong S, Avihingsanon Y, Hansasuta P, Vanichanan J, Townamchai N. Benefits of Switching Mycophenolic Acid to Sirolimus on Serological Response after a SARS-CoV-2 Booster Dose among Kidney Transplant Recipients: A Pilot Study. Vaccines (Basel) 2022; 10:vaccines10101685. [PMID: 36298550 PMCID: PMC9609831 DOI: 10.3390/vaccines10101685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/28/2022] [Accepted: 10/06/2022] [Indexed: 11/08/2022] Open
Abstract
Kidney transplant recipients (KTRs) have a suboptimal immune response to COVID-19 vaccination due to the effects of immunosuppression, mostly mycophenolic acid (MPA). This study investigated the benefits of switching from the standard immunosuppressive regimen (tacrolimus (TAC), MPA, and prednisolone) to a regimen of mammalian target of rapamycin inhibitor (mTORi), TAC and prednisolone two weeks pre- and two weeks post-BNT162b2 booster vaccination. A single-center, opened-label pilot study was conducted in KTRs, who received two doses of ChAdOx-1 and a single dose of BNT162b2. The participants were randomly assigned to continue the standard regimen (control group, n = 14) or switched to a sirolimus (an mTORi), TAC, and prednisolone (switching group, n = 14) regimen two weeks before and two weeks after receiving a booster dose of BNT162b2. The anti-SARS-CoV-2 S antibody level after vaccination in the switching group was significantly greater than the control group (4051.0 [IQR 3142.0-6466.0] BAU/mL vs. 2081.0 [IQR 1077.0-3960.0] BAU/mL, respectively; p = 0.01). One participant who was initially seronegative in the control group remained seronegative after the booster dose. These findings suggest humoral immune response benefits of switching the standard immunosuppressive regimen to the regimen of mTORi, TAC, and prednisolone in KTRs during vaccination.
Collapse
Affiliation(s)
- Athiphat Banjongjit
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
| | - Supitchaya Phirom
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
| | - Jeerath Phannajit
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
- Division of Clinical Epidemiology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Watsamon Jantarabenjakul
- Thai Red Cross Emerging Infectious Diseases Clinical Center, King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
- Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Leilani Paitoonpong
- Thai Red Cross Emerging Infectious Diseases Clinical Center, King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
- Division of Infectious Diseases, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Wonngarm Kittanamongkolchai
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
- Mahachakri Sirindhorn Clinical Research Center, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Renal Immunology and Renal Transplant Research Unit, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Salin Wattanatorn
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
| | | | - Somchai Eiam-Ong
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
| | - Yingyos Avihingsanon
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
- Renal Immunology and Renal Transplant Research Unit, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pokrath Hansasuta
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
| | - Jakapat Vanichanan
- Division of Infectious Diseases, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Natavudh Townamchai
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
- Renal Immunology and Renal Transplant Research Unit, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Excellence Center for Solid Organ Transplantation, King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
- Correspondence:
| |
Collapse
|
36
|
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: 0] [Impact Index Per Article: 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
|
37
|
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
|
38
|
Immunity to a third BNT162B2 COVID-19 vaccine after heart transplantation: Bridging the knowledge gap to end the pandemic for organ transplant recipients. J Heart Lung Transplant 2022; 41:1426-1428. [PMID: 35933295 PMCID: PMC9271413 DOI: 10.1016/j.healun.2022.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 01/31/2023] Open
|
39
|
Kantauskaite M, Müller L, Hillebrandt J, Lamberti J, Fischer S, Kolb T, Ivens K, Koch M, Andree M, Lübke N, Schmitz M, Luedde T, Orth HM, Feldt T, Schaal H, Adams O, Schmidt C, Kittel M, Königshausen E, Rump LC, Timm J, Stegbauer J. Immune response to third SARS-CoV-2 vaccination in seronegative kidney transplant recipients: Possible improvement by mycophenolate mofetil reduction. Clin Transplant 2022; 36:e14790. [PMID: 35997031 PMCID: PMC9539238 DOI: 10.1111/ctr.14790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/07/2022] [Accepted: 08/02/2022] [Indexed: 12/15/2022]
Abstract
Modification of vaccination strategies is necessary to improve the immune response to SARS-CoV-2 vaccination in kidney transplant recipients (KTRs). This multicenter observational study analyzed the effects of the third SARS-CoV-2 vaccination in previously seronegative KTRs with the focus on temporary mycophenolate mofetil (MMF) dose reduction within propensity matched KTRs. 56 out of 174 (32%) previously seronegative KTRs became seropositive after the third vaccination with only three KTRs developing neutralizing antibodies against the omicron variant. Multivariate logistic regression revealed that initial antibody levels, graft function, time after transplantation and MMF trough levels had an influence on seroconversion (P < .05). After controlling for confounders, the effect of MMF dose reduction before the third vaccination was calculated using propensity score matching. KTRs with a dose reduction of ≥33% showed a significant decrease in MMF trough levels to 1.8 (1.2-2.5) μg/ml and were more likely to seroconvert than matched controls (P = .02). Therefore, a MMF dose reduction of 33% or more before vaccination is a promising approach to improve success of SARS-CoV-2 vaccination in KTRs.
Collapse
Affiliation(s)
- Marta Kantauskaite
- Department of NephrologyMedical FacultyUniversity Hospital DüsseldorfHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany
| | - Lisa Müller
- Institute of VirologyMedical FacultyUniversity Hospital DüsseldorfHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany
| | - Jonas Hillebrandt
- Department of NephrologyMedical FacultyUniversity Hospital DüsseldorfHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany
| | - Joshua Lamberti
- Department of NephrologyMedical FacultyUniversity Hospital DüsseldorfHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany
| | - Svenja Fischer
- Department of NephrologyMedical FacultyUniversity Hospital DüsseldorfHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany
| | - Thilo Kolb
- Department of NephrologyMedical FacultyUniversity Hospital DüsseldorfHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany,KfH Kuratorium für Dialyse und Nierentransplantation e.VKfH‐Nierenzentrum Moorenstrasse 5DüsseldorfGermany
| | - Katrin Ivens
- Department of NephrologyMedical FacultyUniversity Hospital DüsseldorfHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany,KfH Kuratorium für Dialyse und Nierentransplantation e.VKfH‐Nierenzentrum Moorenstrasse 5DüsseldorfGermany
| | - Michael Koch
- Medizinisches VersorgungszentrumNephrocare MettmannMettmannGermany
| | - Marcel Andree
- Institute of VirologyMedical FacultyUniversity Hospital DüsseldorfHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany
| | - Nadine Lübke
- Institute of VirologyMedical FacultyUniversity Hospital DüsseldorfHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany
| | - Michael Schmitz
- Department of NephrologyStädtisches Klinikum SolingenSolingenGermany
| | - Tom Luedde
- Department of GastroenterologyHepatology and Infectious diseasesMedical FacultyUniversity Hospital DüsseldorfHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany
| | - Hans Martin Orth
- Department of GastroenterologyHepatology and Infectious diseasesMedical FacultyUniversity Hospital DüsseldorfHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany
| | - Torsten Feldt
- Department of GastroenterologyHepatology and Infectious diseasesMedical FacultyUniversity Hospital DüsseldorfHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany
| | - Heiner Schaal
- Institute of VirologyMedical FacultyUniversity Hospital DüsseldorfHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany
| | - Ortwin Adams
- Institute of VirologyMedical FacultyUniversity Hospital DüsseldorfHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany
| | - Claudia Schmidt
- Department of NephrologyMedical FacultyUniversity Hospital DüsseldorfHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany
| | - Margarethe Kittel
- Department of NephrologyMedical FacultyUniversity Hospital DüsseldorfHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany
| | - Eva Königshausen
- Department of NephrologyMedical FacultyUniversity Hospital DüsseldorfHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany,KfH Kuratorium für Dialyse und Nierentransplantation e.VKfH‐Nierenzentrum Moorenstrasse 5DüsseldorfGermany
| | - Lars C. Rump
- Department of NephrologyMedical FacultyUniversity Hospital DüsseldorfHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany,KfH Kuratorium für Dialyse und Nierentransplantation e.VKfH‐Nierenzentrum Moorenstrasse 5DüsseldorfGermany
| | - Jörg Timm
- Institute of VirologyMedical FacultyUniversity Hospital DüsseldorfHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany
| | - Johannes Stegbauer
- Department of NephrologyMedical FacultyUniversity Hospital DüsseldorfHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany,KfH Kuratorium für Dialyse und Nierentransplantation e.VKfH‐Nierenzentrum Moorenstrasse 5DüsseldorfGermany
| |
Collapse
|
40
|
SARS-CoV-2 Vaccination in Solid-Organ Transplant Recipients. Vaccines (Basel) 2022; 10:vaccines10091430. [PMID: 36146506 PMCID: PMC9503203 DOI: 10.3390/vaccines10091430] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has posed significant global challenges for solid organ transplant (SOT) recipients. Mortality rates of COVID-19 in this patient population remain high, despite new available therapeutic options and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) vaccination. Priority access to SARS-CoV-2 vaccination for waitlisted candidates and for SOT patients and their family members is recommended since the advantage from vaccination reduces the risk of COVID-19-related complications. However, immunogenicity and efficacy of COVID-19 vaccines are lower in waitlisted candidates and SOT recipients than in the general population. Routine systematic assessment of humoral and cellular immune responses after SARS-CoV-2 vaccination is controversial, although highly recommended for investigation and improvement of knowledge. SOT recipients should continue to adhere to preventive protective measures despite vaccination and may undergo passive antibody prophylaxis. This article seeks to provide an update on SARS-CoV-2 vaccination and preventive measures in SOT recipients based on existing literature and international guidelines.
Collapse
|
41
|
Kreuzberger N, Hirsch C, Andreas M, Böhm L, Bröckelmann PJ, Di Cristanziano V, Golinski M, Hausinger RI, Mellinghoff S, Lange B, Lischetzki T, Kappler V, Mikolajewska A, Monsef I, Park YS, Piechotta V, Schmaderer C, Stegemann M, Vanshylla K, Weber F, Weibel S, Stephani C, Skoetz N. Immunity after COVID-19 vaccination in people with higher risk of compromised immune status: a scoping review. Cochrane Database Syst Rev 2022; 8:CD015021. [PMID: 35943061 PMCID: PMC9361430 DOI: 10.1002/14651858.cd015021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND High efficacy in terms of protection from severe COVID-19 has been demonstrated for several SARS-CoV-2 vaccines. However, patients with compromised immune status develop a weaker and less stable immune response to vaccination. Strong immune response may not always translate into clinical benefit, therefore it is important to synthesise evidence on modified schemes and types of vaccination in these population subgroups for guiding health decisions. As the literature on COVID-19 vaccines continues to expand, we aimed to scope the literature on multiple subgroups to subsequently decide on the most relevant research questions to be answered by systematic reviews. OBJECTIVES To provide an overview of the availability of existing literature on immune response and long-term clinical outcomes after COVID-19 vaccination, and to map this evidence according to the examined populations, specific vaccines, immunity parameters, and their way of determining relevant long-term outcomes and the availability of mapping between immune reactivity and relevant outcomes. SEARCH METHODS We searched the Cochrane COVID-19 Study Register, the Web of Science Core Collection, and the World Health Organization COVID-19 Global literature on coronavirus disease on 6 December 2021. SELECTION CRITERIA: We included studies that published results on immunity outcomes after vaccination with BNT162b2, mRNA-1273, AZD1222, Ad26.COV2.S, Sputnik V or Sputnik Light, BBIBP-CorV, or CoronaVac on predefined vulnerable subgroups such as people with malignancies, transplant recipients, people undergoing renal replacement therapy, and people with immune disorders, as well as pregnant and breastfeeding women, and children. We included studies if they had at least 100 participants (not considering healthy control groups); we excluded case studies and case series. DATA COLLECTION AND ANALYSIS We extracted data independently and in duplicate onto an online data extraction form. Data were represented as tables and as online maps to show the frequency of studies for each item. We mapped the data according to study design, country of participant origin, patient comorbidity subgroup, intervention, outcome domains (clinical, safety, immunogenicity), and outcomes. MAIN RESULTS: Out of 25,452 identified records, 318 studies with a total of more than 5 million participants met our eligibility criteria and were included in the review. Participants were recruited mainly from high-income countries between January 2020 and 31 October 2021 (282/318); the majority of studies included adult participants (297/318). Haematological malignancies were the most commonly examined comorbidity group (N = 54), followed by solid tumours (N = 47), dialysis (N = 48), kidney transplant (N = 43), and rheumatic diseases (N = 28, 17, and 15 for mixed diseases, multiple sclerosis, and inflammatory bowel disease, respectively). Thirty-one studies included pregnant or breastfeeding women. The most commonly administered vaccine was BNT162b2 (N = 283), followed by mRNA-1273 (N = 153), AZD1222 (N = 66), Ad26.COV2.S (N = 42), BBIBP-CorV (N = 15), CoronaVac (N = 14), and Sputnik V (N = 5; no studies were identified for Sputnik Light). Most studies reported outcomes after regular vaccination scheme. The majority of studies focused on immunogenicity outcomes, especially seroconversion based on binding antibody measurements and immunoglobulin G (IgG) titres (N = 179 and 175, respectively). Adverse events and serious adverse events were reported in 126 and 54 studies, whilst SARS-CoV-2 infection irrespective of severity was reported in 80 studies. Mortality due to SARS-CoV-2 infection was reported in 36 studies. Please refer to our evidence gap maps for more detailed information. AUTHORS' CONCLUSIONS Up to 6 December 2021, the majority of studies examined data on mRNA vaccines administered as standard vaccination schemes (two doses approximately four to eight weeks apart) that report on immunogenicity parameters or adverse events. Clinical outcomes were less commonly reported, and if so, were often reported as a secondary outcome observed in seroconversion or immunoglobulin titre studies. As informed by this scoping review, two effectiveness reviews (on haematological malignancies and kidney transplant recipients) are currently being conducted.
Collapse
Affiliation(s)
- Nina Kreuzberger
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Caroline Hirsch
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Marike Andreas
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Lena Böhm
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Paul J Bröckelmann
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Max-Planck Institute for the Biology of Ageing, Cologne, Germany
| | - Veronica Di Cristanziano
- Laboratory of Experimental Immunology, Institute of Virology, University Hospital of Cologne, Cologne, Germany
| | - Martin Golinski
- Department of Anesthesiology, University of Goettingen Medical Center, Goettingen, Germany
| | - Renate Ilona Hausinger
- Department of Nephrology, Klinikum rechts der Isar, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Sibylle Mellinghoff
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Germany
| | - Berit Lange
- Department of Epidemiology, Helmholtz Centre for Infection Research, Brunswick, Germany
- Translational Unit BBD, German Center for Infection Research (DZIF), Brunswick, Germany
| | - Tina Lischetzki
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Verena Kappler
- Department of Nephrology, Klinikum rechts der Isar, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Agata Mikolajewska
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Centre for Biological Threats and Special Pathogens (ZBS), Strategy and Incident Response (ZBS7), Clinical Management and Infection Control (ZBS7.1), Robert Koch Institute, Berlin, Germany
| | - Ina Monsef
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Yun Soo Park
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Vanessa Piechotta
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Christoph Schmaderer
- Department of Nephrology, Klinikum rechts der Isar, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Miriam Stegemann
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kanika Vanshylla
- Laboratory of Experimental Immunology, Institute of Virology, University Hospital of Cologne, Cologne, Germany
| | - Florencia Weber
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Stephanie Weibel
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Caspar Stephani
- Department of Anesthesiology, University of Goettingen Medical Center, Goettingen, Germany
| | - Nicole Skoetz
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
42
|
de Nattes T, Candon S, Lemée V, Laurent C, Guerrot D, Bertrand D. Humoral response to a fourth dose of mRNA vaccine in kidney transplant recipients responders to 3 doses of mRNA vaccine. Clin Kidney J 2022; 15:2343-2345. [PMID: 36381362 PMCID: PMC9664567 DOI: 10.1093/ckj/sfac183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Indexed: 12/02/2022] Open
Affiliation(s)
- Tristan de Nattes
- Nephrology - Kidney Transplant Unit, Rouen University Hospital , Rouen , France
- Department of Immunology, and Biotherapies, UNIROUEN, INSERM U1234, Normandy University, Rouen University Hospital , Rouen , France
| | - Sophie Candon
- Department of Immunology, and Biotherapies, UNIROUEN, INSERM U1234, Normandy University, Rouen University Hospital , Rouen , France
| | - Véronique Lemée
- Department of Virology, Rouen University Hospital , Rouen , France
| | - Charlotte Laurent
- Nephrology - Kidney Transplant Unit, Rouen University Hospital , Rouen , France
| | - Dominique Guerrot
- Nephrology - Kidney Transplant Unit, Rouen University Hospital , Rouen , France
| | - Dominique Bertrand
- Nephrology - Kidney Transplant Unit, Rouen University Hospital , Rouen , France
| |
Collapse
|
43
|
Immunity after COVID-19 Recovery and Vaccination: Similarities and Differences. Vaccines (Basel) 2022; 10:vaccines10071068. [PMID: 35891232 PMCID: PMC9322013 DOI: 10.3390/vaccines10071068] [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: 04/26/2022] [Revised: 06/20/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is associated with a robust immune response. The development of systemic inflammation leads to a hyperinflammatory state due to cytokine release syndrome during severe COVID-19. The emergence of many new SARS-CoV-2 variants across the world deteriorates the protective antiviral immunity induced after infection or vaccination. The innate immune response to SARS-CoV-2 is crucial for determining the fate of COVID-19 symptomatology. T cell-mediated immunity is the main factor of the antiviral immune response; moreover, SARS-CoV-2 infection initiates a rapid B-cell response. In this paper, we present the current state of knowledge on immunity after COVID-19 infection and vaccination. We discuss the mechanisms of immune response to various types of vaccines (nucleoside-modified, adenovirus-vectored, inactivated virus vaccines and recombinant protein adjuvanted formulations). This includes specific aspects of vaccination in selected patient populations with altered immune activity (the elderly, children, pregnant women, solid organ transplant recipients, patients with systemic rheumatic diseases or malignancies). We also present diagnostic and research tools available to study the anti-SARS-CoV-2 cellular and humoral immune responses.
Collapse
|
44
|
Opsomer R, Kuypers D. COVID-19 and solid organ transplantation: Finding the right balance. Transplant Rev (Orlando) 2022; 36:100710. [PMID: 35809422 PMCID: PMC9251959 DOI: 10.1016/j.trre.2022.100710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Roxanne Opsomer
- Catholic University of Leuven, Faculty of Medicine, Herestraat 49, 3000 Leuven, Belgium.
| | - Dirk Kuypers
- University Hospitals Leuven, Department of Nephrology and Renal Transplantation; Catholic University Leuven, Department of Microbiology, Immunology and Transplantation, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
45
|
Ronicke S, Osmanodja B, Budde K, Jens A, Hammett C, Koch N, Zukunft B, Bachmann F, Choi M, Weber U, Eberspächer B, Hofmann J, Grunow F, Mikhailov M, Halleck F, Schrezenmeier E. Declining Course of Humoral Immune Response in Initially Responding Kidney Transplant Recipients after Repeated SARS-CoV-2 Vaccination. J Clin Med 2022; 11:3291. [PMID: 35743365 PMCID: PMC9224612 DOI: 10.3390/jcm11123291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 02/06/2023] Open
Abstract
The immunogenicity of SARS-CoV-2 vaccines in kidney transplant recipients is limited, resulting in inadequately low serological response rates and low immunoglobulin (Ig) levels, correlating with reduced protection against death and hospitalization from COVID-19. We retrospectively examined the time course of anti-SARS-CoV-2 Ig antibody levels after up to five repeated vaccinations in 644 previously nonresponding kidney transplant recipients. Using anti SARS-CoV-2 IgG/IgA ELISA and the total Ig ECLIA assays, we compared antibody levels at 1 month with levels at 2 and 4 months, respectively. Additionally, we correlated the measurements of the used assays. Between 1 and 2 months, and between 1 and 4 months, mean anti-SARS-CoV-2 Ig levels in responders decreased by 14% and 25%, respectively, depending on the assay. Absolute Ig values and time course of antibody levels showed high interindividual variability. Ig levels decreased by at least 20% in 77 of 148 paired samples with loss of sufficient serological protection over time occurring in 18 out of 148 (12.2%). IgG ELISA and total Ig ECLIA assays showed a strong positive correlation (Kendall's tau = 0.78), yet the two assays determined divergent results in 99 of 751 (13.2%) measurements. IgG and IgA assays showed overall strong correlation but divergent results in 270 of 1.173 (23.0%) cases and only weak correlation of antibody levels in positive samples. Large interindividual variability and significant loss of serological response after 4 months supports repeated serological sampling and consideration of shorter vaccination intervals in kidney transplant recipients.
Collapse
Affiliation(s)
- Simon Ronicke
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (B.O.); (K.B.); (A.J.); (C.H.); (N.K.); (B.Z.); (F.B.); (M.C.); (U.W.); (F.G.); (M.M.); (F.H.); (E.S.)
| | - Bilgin Osmanodja
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (B.O.); (K.B.); (A.J.); (C.H.); (N.K.); (B.Z.); (F.B.); (M.C.); (U.W.); (F.G.); (M.M.); (F.H.); (E.S.)
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (B.O.); (K.B.); (A.J.); (C.H.); (N.K.); (B.Z.); (F.B.); (M.C.); (U.W.); (F.G.); (M.M.); (F.H.); (E.S.)
| | - Annika Jens
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (B.O.); (K.B.); (A.J.); (C.H.); (N.K.); (B.Z.); (F.B.); (M.C.); (U.W.); (F.G.); (M.M.); (F.H.); (E.S.)
| | - Charlotte Hammett
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (B.O.); (K.B.); (A.J.); (C.H.); (N.K.); (B.Z.); (F.B.); (M.C.); (U.W.); (F.G.); (M.M.); (F.H.); (E.S.)
| | - Nadine Koch
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (B.O.); (K.B.); (A.J.); (C.H.); (N.K.); (B.Z.); (F.B.); (M.C.); (U.W.); (F.G.); (M.M.); (F.H.); (E.S.)
| | - Bianca Zukunft
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (B.O.); (K.B.); (A.J.); (C.H.); (N.K.); (B.Z.); (F.B.); (M.C.); (U.W.); (F.G.); (M.M.); (F.H.); (E.S.)
| | - Friederike Bachmann
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (B.O.); (K.B.); (A.J.); (C.H.); (N.K.); (B.Z.); (F.B.); (M.C.); (U.W.); (F.G.); (M.M.); (F.H.); (E.S.)
| | - Mira Choi
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (B.O.); (K.B.); (A.J.); (C.H.); (N.K.); (B.Z.); (F.B.); (M.C.); (U.W.); (F.G.); (M.M.); (F.H.); (E.S.)
| | - Ulrike Weber
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (B.O.); (K.B.); (A.J.); (C.H.); (N.K.); (B.Z.); (F.B.); (M.C.); (U.W.); (F.G.); (M.M.); (F.H.); (E.S.)
| | | | - Jörg Hofmann
- Labor Berlin—Charité Vivantes GmbH, 13353 Berlin, Germany; (B.E.); (J.H.)
| | - Fritz Grunow
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (B.O.); (K.B.); (A.J.); (C.H.); (N.K.); (B.Z.); (F.B.); (M.C.); (U.W.); (F.G.); (M.M.); (F.H.); (E.S.)
| | - Michael Mikhailov
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (B.O.); (K.B.); (A.J.); (C.H.); (N.K.); (B.Z.); (F.B.); (M.C.); (U.W.); (F.G.); (M.M.); (F.H.); (E.S.)
| | - Fabian Halleck
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (B.O.); (K.B.); (A.J.); (C.H.); (N.K.); (B.Z.); (F.B.); (M.C.); (U.W.); (F.G.); (M.M.); (F.H.); (E.S.)
| | - Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (B.O.); (K.B.); (A.J.); (C.H.); (N.K.); (B.Z.); (F.B.); (M.C.); (U.W.); (F.G.); (M.M.); (F.H.); (E.S.)
- Berlin Institute of Health, Charité—Universitätsmedizin Berlin, BIH Academy, 10117 Berlin, Germany
| |
Collapse
|