1
|
Sangermano M, Negrisolo S, Antoniello B, Vadori M, Cozzi E, Benetti E. Use of Tocilizumab in the treatment of chronic active antibody-mediated rejection in pediatric kidney transplant recipients. Hum Immunol 2024; 85:111088. [PMID: 39146803 DOI: 10.1016/j.humimm.2024.111088] [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: 04/22/2024] [Revised: 06/29/2024] [Accepted: 08/07/2024] [Indexed: 08/17/2024]
Abstract
Chronic active antibody-mediated rejection is one of the leading causes of graft failure and traditional therapies have unclear efficacy. Recent studies suggested that Tocilizumab could stabilize renal function and improve microvascular inflammation. Here we report the outcomes of Tocilizumab therapy in 6 pediatric kidney transplant recipients with biopsy-proven chronic active antibody-mediated rejection resistant to standard treatments. All patients received monthly Tocilizumab infusions for 6 months and were monitored for renal function (creatinine, estimated glomerular filtration rate (eGFR), proteinuria) and Human Leukocyte Antigens (HLA) and non-HLA antibodies at baseline and 3 and 6 months after Tocilizumab initiation. For each patient, a follow-up biopsy was scheduled at the end of the treatment. Renal function did not show stabilization or improvement (mean eGFR 37 ml/min/1.73 m2 pre-Tocilizumab and 27 ml/min/1.73 m2 3 months after-Tocilizumab) and proteinuria remained stable. Moreover, Tocilizumab had no impact on HLA and non-HLA antibodies. Graft loss was observed in 3 patients (50 %) and 4 patients who underwent post-treatment biopsy showed a worsening in overall chronicity scores. In our pediatric series, rescue therapy with Tocilizumab did not appear to be effective in modifying the natural history of chronic active antibody-mediated rejection.
Collapse
Affiliation(s)
- Maria Sangermano
- Pediatric Nephrology, Department of Women's and Children's Health, Padua University Hospital, Padua, Italy
| | - Susanna Negrisolo
- Laboratory of Immunopathology and Molecular Biology of the Kidney, Department of Women's and Children's Health, Padua University Hospital, Padua, Italy
| | - Benedetta Antoniello
- Laboratory of Immunopathology and Molecular Biology of the Kidney, Department of Women's and Children's Health, Padua University Hospital, Padua, Italy
| | - Marta Vadori
- Transplant Immunology Unit, Department of CardioThoraco-Vascular Sciences and Public Health, Padua University Hospital, Padua, Italy
| | - Emanuele Cozzi
- Transplant Immunology Unit, Department of CardioThoraco-Vascular Sciences and Public Health, Padua University Hospital, Padua, Italy
| | - Elisa Benetti
- Pediatric Nephrology, Department of Women's and Children's Health, Padua University Hospital, Padua, Italy; Laboratory of Immunopathology and Molecular Biology of the Kidney, Department of Women's and Children's Health, Padua University Hospital, Padua, Italy.
| |
Collapse
|
2
|
Diebold M, Mayer KA, Hidalgo L, Kozakowski N, Budde K, Böhmig GA. Chronic Rejection After Kidney Transplantation. Transplantation 2024:00007890-990000000-00858. [PMID: 39192468 DOI: 10.1097/tp.0000000000005187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
In kidney transplantation, ongoing alloimmune processes-commonly triggered by HLA incompatibilities-can trigger chronic transplant rejection, affecting the microcirculation and the tubulointerstitium. Continuous inflammation may lead to progressive, irreversible graft injury, culminating in graft dysfunction and accelerated transplant failure. Numerous experimental and translational studies have delineated a complex interplay of different immune mechanisms driving rejection, with antibody-mediated rejection (AMR) being an extensively studied rejection variant. In microvascular inflammation, a hallmark lesion of AMR, natural killer (NK) cells have emerged as pivotal effector cells. Their essential role is supported by immunohistologic evidence, bulk and spatial transcriptomics, and functional genetics. Despite significant research efforts, a substantial unmet need for approved rejection therapies persists, with many trials yielding negative outcomes. However, several promising therapies are currently under investigation, including felzartamab, a monoclonal antibody targeting the surface molecule CD38, which is highly expressed in NK cells and antibody-producing plasma cells. In an exploratory phase 2 trial in late AMR, this compound has demonstrated potential in resolving molecular and morphologic rejection activity and injury, predominantly by targeting NK cell effector function. These findings inspire hope for effective treatments and emphasize the necessity of further pivotal trials focusing on chronic transplant rejection.
Collapse
Affiliation(s)
- Matthias Diebold
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Katharina A Mayer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Luis Hidalgo
- HLA Laboratory, Division of Transplantation, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | | | - Klemens Budde
- Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Georg A Böhmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
3
|
Akifova A, Budde K, Oellerich M, Beck J, Bornemann-Kolatzki K, Schütz E, Osmanodja B. Perspective for Donor-Derived Cell-Free DNA in Antibody-Mediated Rejection After Kidney Transplantation: Defining Context of Use and Clinical Implications. Transpl Int 2024; 37:13239. [PMID: 39188271 PMCID: PMC11345135 DOI: 10.3389/ti.2024.13239] [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: 05/10/2024] [Accepted: 07/30/2024] [Indexed: 08/28/2024]
Abstract
Antibody-mediated rejection (AMR) is a major cause of graft failure limiting long-term graft survival after kidney transplantation. Current diagnostic strategy to detect AMR is suboptimal and requires further improvement. Previously suggested treatment regimens for AMR could not demonstrate efficacy, however novel therapeutic agents are currently under investigation. Donor-derived cell-free DNA (dd-cfDNA) is a novel non-invasive biomarker for allograft injury, that has been mainly studied in the context of rejection. Its short-half-life in circulation and injury-dependent release are its key advantages that contribute to its superior diagnostic accuracy, compared to traditional biomarkers. Moreover, previous studies showed that dd-cfDNA-release is well-linked to histological and molecular features of AMR, and thus able to reflect real-time injury. Further observations suggest that dd-cfDNA can be used as a suitable screening tool for early detection of AMR in patients with donor-specific-anti-HLA-antibodies (DSA), as well as for monitoring AMR activity after anti-rejection treatment. The weight of evidence suggests that the integration of dd-cfDNA in the graft surveillance of patients with AMR, or those suspicious of AMR (e.g., due to the presence of donor-specific anti-HLA-antibodies) has an added value and might have a positive impact on outcomes in this specific cohort.
Collapse
Affiliation(s)
- Aylin Akifova
- Department of Nephrology and Intensive Care, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Intensive Care, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Oellerich
- Department of Clinical Pharmacology, University Medical Center Göttingen, Göttingen, Germany
| | - Julia Beck
- Chronix Biomedical GmbH, Göttingen, Germany
| | | | | | - Bilgin Osmanodja
- Department of Nephrology and Intensive Care, Charité—Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
4
|
Osmanodja B, Akifova A, Budde K, Oellerich M, Beck J, Bornemann-Kolatzki K, Schütz E, Velden J, Lehmann C, Krüger BM, Bachmann A, Kowald J. Donor-Derived Cell-Free DNA as a Companion Biomarker for AMR Treatment With Daratumumab: Case Series. Transpl Int 2024; 37:13213. [PMID: 39149569 PMCID: PMC11325154 DOI: 10.3389/ti.2024.13213] [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: 04/30/2024] [Accepted: 07/22/2024] [Indexed: 08/17/2024]
Abstract
Antibody-mediated rejection (AMR) is among the most frequent causes for graft loss after kidney transplantation. While there are no approved therapies, several case reports with daratumumab and the very recent phase 2 trial of felzartamab in AMR have indicated the potential efficacy of therapeutic interventions targeting CD38. Donor-derived cell-free DNA (dd-cfDNA) is an emerging biomarker with injury-specific release and a short half-life, which could facilitate early diagnosis of AMR and monitoring of treatment response. We describe two cases of patients with chronic active AMR, who were treated with monthly daratumumab infusions, and in whom donor-derived cell-free DNA (dd-cfDNA) was measured longitudinally to monitor treatment response. In both patients, daratumumab treatment led to stabilization of kidney function parameters, a strong decline of dd-cfDNA below the previously established threshold for rejection, and partial or complete histologic resolution of AMR activity. Our case series suggests that dd-cfDNA may be a useful companion biomarker for longitudinal monitoring of anti-CD38 treatment in patients with AMR.
Collapse
Affiliation(s)
- Bilgin Osmanodja
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Aylin Akifova
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Michael Oellerich
- Department of Clinical Pharmacology, University Medical Center Göttingen, Göttingen, Germany
| | - Julia Beck
- Chronix Biomedical GmbH, Göttingen, Germany
| | | | | | - Joachim Velden
- Department of Nephropathology, Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Claudia Lehmann
- Institute for Transfusion Medicine, Laboratory for Transplantation Immunology, University Hospital Leipzig, Leipzig, Germany
| | - Bastian Malte Krüger
- Medical Department III, Division of Nephrology, University of Leipzig Medical Center, Leipzig, Germany
| | - Anette Bachmann
- Medical Department III, Division of Nephrology, University of Leipzig Medical Center, Leipzig, Germany
| | - Jan Kowald
- Medical Department III, Division of Nephrology, University of Leipzig Medical Center, Leipzig, Germany
| |
Collapse
|
5
|
Nellore A, Zumaquero E, Seifert M. T-bet + B Cells in Humans: Protective and Pathologic Functions. Transplantation 2024; 108:1709-1714. [PMID: 38051131 PMCID: PMC11150333 DOI: 10.1097/tp.0000000000004889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 12/07/2023]
Abstract
The humoral immune system comprises B cells and plasma cells, which play important roles in organ transplantation, ranging from the production of both protective and injurious antibodies as well as cytokines that can promote operational tolerance. Recent data from conditions outside of transplantation have identified a novel human B-cell subset that expresses the transcription factor T-bet and exerts pleiotropic functions by disease state. Here, we review the generation, activation, and functions of the T-bet + B-cell subset outside of allotransplantation, and consider the relevance of this subset as mediators of allograft injury.
Collapse
Affiliation(s)
- Anoma Nellore
- Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, AL
| | - Esther Zumaquero
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL
| | - Michael Seifert
- Division of Pediatric Nephrology, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
6
|
Killian JT, Glenn King R, Lucander ACK, Kizziah JL, Fucile CF, Diaz-Avalos R, Qiu S, Silva-Sanchez A, Mousseau BJ, Macon KJ, Callahan AR, Yang G, Emon Hossain M, Akther J, Good DB, Kelso S, Houp JA, Rosenblum F, Porrett PM, Ong SC, Kumar V, Saphire EO, Kearney JF, Randall TD, Rosenberg AF, Green TJ, Lund FE. HLA topography enforces shared and convergent immunodominant B cell and antibody alloresponses in transplant recipients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.31.534734. [PMID: 37034637 PMCID: PMC10081326 DOI: 10.1101/2023.03.31.534734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Donor-specific antibody (DSA) responses against human leukocyte antigen (HLA) proteins mismatched between kidney transplant donors and recipients cause allograft loss. The rules governing the immunogenicity of non-self donor HLA are poorly understood. Using single-cell, molecular, structural, and proteomic techniques, we profiled the HLA-specific B cell response in the kidney and blood of a transplant recipient with antibody-mediated rejection (AMR). We observed an immunodominant B cell antibody response focused on topographically exposed, solvent-accessible mismatched HLA residues along the peptide-binding groove - a subregion comprising only 20% of the HLA molecule. We further demonstrated that, even within a diverse cohort of transplant recipients, the B cell alloresponse consistently converges on this same immunodominant subregion on the crown of the HLA molecule. Based on these findings, we propose that B cell immunodominance in transplant rejection relies on antigenic topography, and we suggest that this link could be exploited for organ matching and therapeutics.
Collapse
|
7
|
Namba-Hamano T, Hamano T, Doi Y, Hiraoka A, Yonishi H, Sakai S, Takahashi A, Mizui M, Nakazawa S, Yamanaka K, Kakuta Y, Imamura R, Nonomura N, Isaka Y. Clinical Impacts of Allograft Biopsy in Renal Transplant Recipients 10 Years or Longer After Transplantation. Transpl Int 2024; 37:13022. [PMID: 39091613 PMCID: PMC11292417 DOI: 10.3389/ti.2024.13022] [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: 03/22/2024] [Accepted: 06/25/2024] [Indexed: 08/04/2024]
Abstract
We aimed to investigate the clinical value of allograft biopsy performed long after renal transplantation. We retrospectively evaluated 99 allograft biopsies in recipients with transplantation vintages of 10 years or longer. Mixed-effects model showed that 1-year estimated glomerular filtration rate (eGFR) slopes after biopsy were significantly greater than those before biopsy [-3.13, -4.42 mL/min/1.73 m2/year, p = 0.01]. Renal biopsy changed the treatment strategies in more than half of the patients. Improvement in eGFR slopes was pronounced in 51 patients with treatment modification based on the biopsy results [2.27 (95% confidence interval (CI): 0.66, 3.89) mL/min/1.73 m2/year], whereas no improvement was observed in those without [0.33 (95% CI: -1.05, 1.71) mL/min/1.73 m2/year, Pinteraction = 0.001]. Among the treatment modifications, enhancement of immunosuppression (IS) led to the most remarkable improvement in eGFR slope. Patients with g scores ≥2 were more likely to receive IS enhancement than those with g scores = 0 [odds ratio; 15.0 (95% CI: 1.65, 136)]. Patients with active glomerulitis (g ≥ 1) without chronicity (cg ≤ 1) showed the most significant improvement in eGFR slope. Given the prevalence of active glomerulitis (g ≥ 1, 21%), which is responsive to treatment even long after transplantation, and the observed magnitude of eGFR slope improvement, renal biopsy can indeed improve allograft prognosis.
Collapse
Affiliation(s)
- Tomoko Namba-Hamano
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Takayuki Hamano
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Nephrology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yohei Doi
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Atsuko Hiraoka
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hiroaki Yonishi
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shinsuke Sakai
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Atsushi Takahashi
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Masayuki Mizui
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shigeaki Nakazawa
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kazuaki Yamanaka
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yoichi Kakuta
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Ryoichi Imamura
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
8
|
Mayer KA, Schrezenmeier E, Diebold M, Halloran PF, Schatzl M, Schranz S, Haindl S, Kasbohm S, Kainz A, Eskandary F, Doberer K, Patel UD, Dudani JS, Regele H, Kozakowski N, Kläger J, Boxhammer R, Amann K, Puchhammer-Stöckl E, Vietzen H, Beck J, Schütz E, Akifova A, Firbas C, Gilbert HN, Osmanodja B, Halleck F, Jilma B, Budde K, Böhmig GA. A Randomized Phase 2 Trial of Felzartamab in Antibody-Mediated Rejection. N Engl J Med 2024; 391:122-132. [PMID: 38804514 DOI: 10.1056/nejmoa2400763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
BACKGROUND Antibody-mediated rejection is a leading cause of kidney-transplant failure. The targeting of CD38 to inhibit graft injury caused by alloantibodies and natural killer (NK) cells may be a therapeutic option. METHODS In this phase 2, double-blind, randomized, placebo-controlled trial, we assigned patients with antibody-mediated rejection that had occurred at least 180 days after transplantation to receive nine infusions of the CD38 monoclonal antibody felzartamab (at a dose of 16 mg per kilogram of body weight) or placebo for 6 months, followed by a 6-month observation period. The primary outcome was the safety and side-effect profile of felzartamab. Key secondary outcomes were renal-biopsy results at 24 and 52 weeks, donor-specific antibody levels, peripheral NK-cell counts, and donor-derived cell-free DNA levels. RESULTS A total of 22 patients underwent randomization (11 to receive felzartamab and 11 to receive placebo). The median time from transplantation until trial inclusion was 9 years. Mild or moderate infusion reactions occurred in 8 patients in the felzartamab group. Serious adverse events occurred in 1 patient in the felzartamab group and in 4 patients in the placebo group; graft loss occurred in 1 patient in the placebo group. At week 24, resolution of morphologic antibody-mediated rejection was more frequent with felzartamab (in 9 of 11 patients [82%]) than with placebo (in 2 of 10 patients [20%]), for a difference of 62 percentage points (95% confidence interval [CI], 19 to 100) and a risk ratio of 0.23 (95% confidence interval [CI], 0.06 to 0.83). The median microvascular inflammation score was lower in the felzartamab group than in the placebo group (0 vs. 2.5), for a mean difference of -1.95 (95% CI, -2.97 to -0.92). Also lower was a molecular score reflecting the probability of antibody-mediated rejection (0.17 vs. 0.77) and the level of donor-derived cell-free DNA (0.31% vs. 0.82%). At week 52, the recurrence of antibody-mediated rejection was reported in 3 of 9 patients who had a response to felzartamab, with an increase in molecular activity and biomarker levels toward baseline levels. CONCLUSIONS Felzartamab had acceptable safety and side-effect profiles in patients with antibody-mediated rejection. (Funded by MorphoSys and Human Immunology Biosciences; ClinicalTrials.gov number, NCT05021484; and EUDRACT number, 2021-000545-40.).
Collapse
Affiliation(s)
- Katharina A Mayer
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Eva Schrezenmeier
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Matthias Diebold
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Philip F Halloran
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Martina Schatzl
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Sabine Schranz
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Susanne Haindl
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Silke Kasbohm
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Alexander Kainz
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Farsad Eskandary
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Konstantin Doberer
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Uptal D Patel
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Jaideep S Dudani
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Heinz Regele
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Nicolas Kozakowski
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Johannes Kläger
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Rainer Boxhammer
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Kerstin Amann
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Elisabeth Puchhammer-Stöckl
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Hannes Vietzen
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Julia Beck
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Ekkehard Schütz
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Aylin Akifova
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Christa Firbas
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Houston N Gilbert
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Bilgin Osmanodja
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Fabian Halleck
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Bernd Jilma
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Klemens Budde
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Georg A Böhmig
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| |
Collapse
|
9
|
Pham JPA, Coronel MM. Unlocking Transplant Tolerance with Biomaterials. Adv Healthc Mater 2024:e2400965. [PMID: 38843866 DOI: 10.1002/adhm.202400965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/31/2024] [Indexed: 07/04/2024]
Abstract
For patients suffering from organ failure due to injury or autoimmune disease, allogeneic organ transplantation with chronic immunosuppression is considered the god standard in terms of clinical treatment. However, the true "holy grail" of transplant immunology is operational tolerance, in which the recipient exhibits a sustained lack of alloreactivity toward unencountered antigen presented by the donor graft. This outcome is resultant from critical changes to the phenotype and genotype of the immune repertoire predicated by the activation of specific signaling pathways responsive to soluble and mechanosensitive cues. Biomaterials have emerged as a medium for interfacing with and reprogramming these endogenous pathways toward tolerance in precise, minimally invasive, and spatiotemporally defined manners. By viewing seminal and contemporary breakthroughs in transplant tolerance induction through the lens of biomaterials-mediated immunomodulation strategies-which include intrinsic material immunogenicity, the depot effect, graft coatings, induction and delivery of tolerogenic immune cells, biomimicry of tolerogenic immune cells, and in situ reprogramming-this review emphasizes the stunning diversity of approaches in the field and spotlights exciting future directions for research to come.
Collapse
Affiliation(s)
- John-Paul A Pham
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Elizabeth Caswell Diabetes Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - María M Coronel
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Elizabeth Caswell Diabetes Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
10
|
Berger M, Baliker M, Van Gelder T, Böhmig GA, Mannon RB, Kumar D, Chadban S, Nickerson P, Lee LA, Djamali A. Chronic Active Antibody-mediated Rejection: Opportunity to Determine the Role of Interleukin-6 Blockade. Transplantation 2024; 108:1109-1114. [PMID: 37941113 PMCID: PMC11042519 DOI: 10.1097/tp.0000000000004822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/17/2023] [Accepted: 08/16/2023] [Indexed: 11/10/2023]
Abstract
Chronic active antibody-mediated rejection (caAMR) is arguably the most important cause of late kidney allograft failure. However, there are no US Food and Drug Administration (FDA)-approved treatments for acute or chronic AMR and there is no consensus on effective treatment. Many trials in transplantation have failed because of slow and/or inadequate enrollment, and no new agent has been approved by the FDA for transplantation in over a decade. Several lines of evidence suggest that interleukin-6 is an important driver of AMR, and clazakizumab, a humanized monoclonal antibody that neutralizes interleukin-6, has shown promising results in phase 2 studies. The IMAGINE trial (Interleukin-6 Blockade Modifying Antibody-mediated Graft Injury and Estimated Glomerular Filtration Rate Decline) (NCT03744910) is the first to be considered by the FDA using a reasonably likely surrogate endpoint (slope of estimated glomerular filtration rate decline >1 y) for accelerated approval and is the only ongoing clinical trial for the treatment of chronic rejection. This trial offers us the opportunity to advance the care for our patients in need, and this article is a call to action for all transplant providers caring for patients with caAMR.
Collapse
Affiliation(s)
- Mel Berger
- Departments of Pediatrics and Pathology, Case Western Reserve University, Cleveland, OH
| | | | - Teun Van Gelder
- Department Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, the Netherlands
| | - Georg A. Böhmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Roslyn B. Mannon
- Division of Nephrology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Deepali Kumar
- Department of Medicine, Division of Transplant Infectious Disease, Ajmera Transplant Centre, Toronto, ON, Canada
| | - Steve Chadban
- Department of Renal Medicine, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Peter Nickerson
- Department of Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Laurie A. Lee
- Research and Development, Transplant Therapeutic Area, CSL Behring, King of Prussia, Pennsylvania, PA
| | - Arjang Djamali
- Department of Medicine, Maine Medical Center, Portland, ME
| |
Collapse
|
11
|
Han F, Shi X, Liao T, Zhang W, Ma M, Leng Q, Jiang W, Na N, Miao Y, Huang Z. Bruton's tyrosine kinase ablation inhibits B cell responses and antibody production for the prevention of chronic rejection in cardiac transplantation. Clin Immunol 2024; 261:109941. [PMID: 38365047 DOI: 10.1016/j.clim.2024.109941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 02/12/2024] [Indexed: 02/18/2024]
Abstract
Chronic rejection is the primary cause of late allograft failure, however, the current treatments for chronic rejection have not yielded desirable therapeutic effects. B cell activation and donor-specific antibody (DSA) production are the primary factors leading to chronic rejection. Bruton's tyrosine kinase (BTK) plays a key role in the activation and differentiation of B cells and in antibody production. This study investigated the efficacy of blocking BTK signalling in the prevention of chronic rejection. BTK signalling was blocked using the BTK inhibitor ibrutinib and gene knockout. In vitro assays were conducted to examine the consequences and underlying mechanisms of BTK blockade in regards to B cell activation, differentiation, and antibody secretion. Additionally, we established a cardiac transplantation mouse model of chronic rejection to explore the preventive effects and mechanisms of BTK ablation on chronic rejection. Ablating BTK signalling in vitro resulted in the inhibition of B cell activation, differentiation, and antibody production. In vivo experiments provided evidence that ablating BTK signalling alleviated chronic rejection, leading to reduced damage in myocardial tissue, neointimal hyperplasia, interstitial fibrosis, inflammatory cell infiltration, and C4d deposition. Allograft survival was prolonged, and B cell responses and DSA production were inhibited as a result. We confirmed that ablation of BTK signalling inhibited B cell response by blocking downstream PLCγ2 phosphorylation and inhibiting the NF-κB, NFAT, and ERK pathways. Our findings demonstrated that ablation of BTK signalling inhibited B cell activation and differentiation, reduced DSA production, and effectively prevented chronic rejection.
Collapse
Affiliation(s)
- Fei Han
- Organ Transplantation Research Institution, Division of Kidney Transplantation, Department of Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaoyi Shi
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Tao Liao
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Zhang
- Organ Transplantation Research Institution, Division of Kidney Transplantation, Department of Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Maolin Ma
- Organ Transplantation Research Institution, Division of Kidney Transplantation, Department of Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qianghua Leng
- Organ Transplantation Research Institution, Division of Kidney Transplantation, Department of Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Weichen Jiang
- Organ Transplantation Research Institution, Division of Kidney Transplantation, Department of Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ning Na
- Organ Transplantation Research Institution, Division of Kidney Transplantation, Department of Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yun Miao
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Zhengyu Huang
- Organ Transplantation Research Institution, Division of Kidney Transplantation, Department of Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
12
|
Streichart L, Felldin M, Ekberg J, Mjörnstedt L, Lindnér P, Lennerling A, Bröcker V, Mölne J, Holgersson J, Daenen K, Wennberg L, Lorant T, Baid-Agrawal S. Tocilizumab in chronic active antibody-mediated rejection: rationale and protocol of an in-progress randomized controlled open-label multi-center trial (INTERCEPT study). Trials 2024; 25:213. [PMID: 38519988 PMCID: PMC10958896 DOI: 10.1186/s13063-024-08020-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/26/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Chronic active antibody-mediated rejection (caAMR) in kidney transplants is associated with irreversible tissue damage and a leading cause of graft loss in the long-term. However, the treatment for caAMR remains a challenge to date. Recently, tocilizumab, a recombinant humanized monoclonal antibody directed against the human interleukin-6 (IL-6) receptor, has shown promise in the treatment of caAMR. However, it has not been systematically investigated so far underscoring the need for randomized controlled studies in this area. METHODS The INTERCEPT study is an investigator-driven randomized controlled open-label multi-center trial in kidney transplant recipients to assess the efficacy of tocilizumab in the treatment of biopsy-proven caAMR. A total of 50 recipients with biopsy-proven caAMR at least 12 months after transplantation will be randomized to receive either tocilizumab (n = 25) added to our standard of care (SOC) maintenance treatment or SOC alone (n = 25) for a period of 24 months. Patients will be followed for an additional 12 months after cessation of study medication. After the inclusion biopsies at baseline, protocol kidney graft biopsies will be performed at 12 and 24 months. The sample size calculation assumed a difference of 5 ml/year in slope of estimated glomerular filtration rate (eGFR) between the two groups for 80% power at an alpha of 0.05. The primary endpoint is the slope of eGFR at 24 months after start of treatment. The secondary endpoints include assessment of the following at 12, 24, and 36 months: composite risk score iBox, safety, evolution and characteristics of donor-specific antibodies (DSA), graft histology, proteinuria, kidney function assessed by measured GFR (mGFR), patient- and death-censored graft survival, and patient-reported outcomes that include transplant-specific well-being, adherence to immunosuppressive medications and perceived threat of the risk of graft rejection. DISCUSSION No effective treatment exists for caAMR at present. Based on the hypothesis that inhibition of IL-6 receptor by tocilizumab will reduce antibody production and reduce antibody-mediated damage, our randomized trial has a potential to provide evidence for a novel treatment strategy for caAMR, therewith slowing the decline in graft function in the long-term. TRIAL REGISTRATION ClinicalTrials.gov NCT04561986. Registered on September 24, 2020.
Collapse
Affiliation(s)
- Lillian Streichart
- Transplant Institute, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Marie Felldin
- Transplant Institute, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Jana Ekberg
- Transplant Institute, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Lars Mjörnstedt
- Transplant Institute, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Per Lindnér
- Transplant Institute, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Annette Lennerling
- Transplant Institute, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Verena Bröcker
- Department of Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Johan Mölne
- Department of Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jan Holgersson
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg and Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kristien Daenen
- Department of Nephrology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lars Wennberg
- Department of Transplantation Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Tomas Lorant
- Section of Transplantation Surgery, Department of Surgical Sciences, Uppsala University Hospital, Uppsala, Sweden
| | - Seema Baid-Agrawal
- Transplant Institute, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, 413 45, Gothenburg, Sweden.
| |
Collapse
|
13
|
Désy O, Béland S, Thivierge MP, Marcoux M, Desgagnés JS, Bouchard-Boivin F, Gama A, Riopel J, Latulippe E, De Serres SA. T follicular helper cells expansion in transplant recipients correlates with graft infiltration and adverse outcomes. Front Immunol 2024; 15:1275933. [PMID: 38384450 PMCID: PMC10879567 DOI: 10.3389/fimmu.2024.1275933] [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: 08/10/2023] [Accepted: 01/24/2024] [Indexed: 02/23/2024] Open
Abstract
Introduction The process of immunization following vaccination in humans bears similarities to that of immunization with allografts. Whereas vaccination aims to elicit a rapid response, in the transplant recipient, immunosuppressants slow the immunization to alloantigens. The induction of CD4+CXCR5+ T follicular helper (Tfh) cells has been shown to correlate with the success of vaccine immunization. Method We studied a cohort of 65 transplant recipients who underwent histological evaluation concurrent with PBMC isolation and follow-up sampling to investigate the phenotypic profiles in the blood and allotissue and analyze their association with clinical events. Results The proportion of circulating Tfh cells was heterogeneous over time. Patients in whom this compartment increased had lower CCR7-PD1+CD4+CXCR5+ T cells during follow-up. These patients exhibited more alloreactive CD4+ T cells using HLA-DR-specific tetramers and a greater proportion of detectable circulating plasmablasts than the controls. Examination of baseline biopsies revealed that expansion of the circulating Tfh compartment did not follow prior intragraft leukocyte infiltration. However, multicolor immunofluorescence microscopy of the grafts showed a greater proportion of CXCR5+ T cells than in the controls. CD4+CXCR5+ cells were predominantly PD1+ and were in close contact with B cells in situ. Despite clinical stability at baseline, circulating Tfh expansion was associated with a higher risk of a composite of anti-HLA donor-specific antibodies, rejection, lower graft function, or graft loss. Conclusion In otherwise stable patients post-transplant, circulating Tfh expansion can identify ongoing alloreactivity, detectable before allograft injury. Tfh expansion is relevant clinically because it predicts poor graft prognosis. These findings have implications for immune surveillance.
Collapse
Affiliation(s)
- Olivier Désy
- Transplantation Unit, Renal Division, Department of Medicine, University Health Center of Quebec, Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Stéphanie Béland
- Transplantation Unit, Renal Division, Department of Medicine, University Health Center of Quebec, Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Marie-Pier Thivierge
- Transplantation Unit, Renal Division, Department of Medicine, University Health Center of Quebec, Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Meagan Marcoux
- Transplantation Unit, Renal Division, Department of Medicine, University Health Center of Quebec, Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Jean-Simon Desgagnés
- Transplantation Unit, Renal Division, Department of Medicine, University Health Center of Quebec, Faculty of Medicine, Laval University, Québec, QC, Canada
| | - François Bouchard-Boivin
- Transplantation Unit, Renal Division, Department of Medicine, University Health Center of Quebec, Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Alcino Gama
- Transplantation Unit, Renal Division, Department of Medicine, University Health Center of Quebec, Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Julie Riopel
- Pathology Division, Department of Medicine, University Health Center of Quebec, Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Eva Latulippe
- Pathology Division, Department of Medicine, University Health Center of Quebec, Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Sacha A. De Serres
- Transplantation Unit, Renal Division, Department of Medicine, University Health Center of Quebec, Faculty of Medicine, Laval University, Québec, QC, Canada
| |
Collapse
|
14
|
Banno T, Hirai T, Oki R, Yagisawa T, Unagami K, Kanzawa T, Omoto K, Shimizu T, Ishida H, Takagi T. Higher Donor Age and Severe Microvascular Inflammation Are Risk Factors for Chronic Rejection After Treatment of Active Antibody-Mediated Rejection. Transpl Int 2024; 37:11960. [PMID: 38371907 PMCID: PMC10869508 DOI: 10.3389/ti.2024.11960] [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: 08/24/2023] [Accepted: 01/10/2024] [Indexed: 02/20/2024]
Abstract
Recent developments in intensive desensitization protocols have enabled kidney transplantation in human leukocyte antigen (HLA)-sensitized recipients. However, cases of active antibody-mediated rejection (AABMR), when they occur, are difficult to manage, graft failure being the worst-case scenario. We aimed to assess the impact of our desensitization and AABMR treatment regimen and identify risk factors for disease progression. Among 849 patients who underwent living-donor kidney transplantation between 2014 and 2021 at our institution, 59 were diagnosed with AABMR within 1 year after transplantation. All patients received combination therapy consisting of steroid pulse therapy, intravenous immunoglobulin, rituximab, and plasmapheresis. Multivariable analysis revealed unrelated donors and preformed donor-specific antibodies as independent risk factors for AABMR. Five-year death-censored graft survival rate was not significantly different between patients with and without AABMR although 27 of 59 patients with AABMR developed chronic AABMR (CABMR) during the study period. Multivariate Cox proportional hazard regression analysis revealed that a donor age greater than 59 years and microvascular inflammation (MVI) score (g + ptc) ≥4 at AABMR diagnosis were independent risk factors for CABMR. Our combination therapy ameliorated AABMR; however, further treatment options should be considered to prevent CABMR, especially in patients with old donors and severe MVI.
Collapse
Affiliation(s)
- Taro Banno
- Department of Urology, Tokyo Women’s Medical University Hospital, Tokyo, Japan
| | - Toshihito Hirai
- Department of Urology, Tokyo Women’s Medical University Hospital, Tokyo, Japan
| | - Rikako Oki
- Department of Organ Transplant Medicine, Tokyo Women’s Medical University Hospital, Tokyo, Japan
| | - Takafumi Yagisawa
- Department of Urology, Tokyo Women’s Medical University Hospital, Tokyo, Japan
| | - Kohei Unagami
- Department of Organ Transplant Medicine, Tokyo Women’s Medical University Hospital, Tokyo, Japan
| | - Taichi Kanzawa
- Department of Urology, Tokyo Women’s Medical University Hospital, Tokyo, Japan
| | - Kazuya Omoto
- Department of Urology, Tokyo Women’s Medical University Hospital, Tokyo, Japan
| | - Tomokazu Shimizu
- Department of Organ Transplant Medicine, Tokyo Women’s Medical University Hospital, Tokyo, Japan
| | - Hideki Ishida
- Department of Organ Transplant Medicine, Tokyo Women’s Medical University Hospital, Tokyo, Japan
| | - Toshio Takagi
- Department of Urology, Tokyo Women’s Medical University Hospital, Tokyo, Japan
| |
Collapse
|
15
|
DiToro D, Murakami N, Pillai S. T-B Collaboration in Autoimmunity, Infection, and Transplantation. Transplantation 2024; 108:386-398. [PMID: 37314442 PMCID: PMC11345790 DOI: 10.1097/tp.0000000000004671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
We have attempted here to provide an up-to-date review of the collaboration between helper T cells and B cells in response to protein and glycoprotein antigens. This collaboration is essential as it not only protects from many pathogens but also contributes to a litany of autoimmune and immune-mediated diseases.
Collapse
Affiliation(s)
- Daniel DiToro
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
| | - Naoka Murakami
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Shiv Pillai
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA
| |
Collapse
|
16
|
De Martin E, McCaughan GW. Antibody-mediated rejection in liver transplant recipients: Some questions, some answers. Liver Transpl 2023; 29:1260-1261. [PMID: 37548557 DOI: 10.1097/lvt.0000000000000239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 07/31/2023] [Indexed: 08/08/2023]
Affiliation(s)
| | - Geoffrey W McCaughan
- Liver Injury and Cancer Program, The Centenary Institute, University of Sydney and Royal Prince Alfred Hospital, Sydney, NSW, Australia
- Australian National Liver Transplant Unit, University of Sydney and Royal Prince Alfred Hospital, Sydney, NSW, Australia
| |
Collapse
|
17
|
Hou YB, Chang S, Chen S, Zhang WJ. Intravenous immunoglobulin in kidney transplantation: Mechanisms of action, clinical applications, adverse effects, and hyperimmune globulin. Clin Immunol 2023; 256:109782. [PMID: 37742791 DOI: 10.1016/j.clim.2023.109782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/11/2023] [Accepted: 09/20/2023] [Indexed: 09/26/2023]
Abstract
Intravenous immunoglobulin (IVIG) has been developed for over 40 years. The mechanisms of action of IVIG are complex and diverse, and there may be multiple mechanisms that combine to influence it. IVIG has been used in kidney transplantation for desensitization, treatment of antibody-mediated rejection, and ABO-incompatible transplantation. and treatment or prevention of some infectious diseases. Hyperimmune globulins such as cytomegalovirus hyperimmune globulin (CMV-IG) and hepatitis B hyperimmune globulin (HBIG) have also been used to protect against cytomegalovirus and hepatitis B virus, respectively. However, IVIG is also associated with some rare but serious adverse effects and some application risks, and clinicians need to weigh the pros and cons and develop individualized treatment programs to benefit more patients. This review will provide an overview of the multiple mechanisms of action, clinical applications, adverse effects, and prophylactic measures of IVIG, and hyperimmune globulin will also be introduced in it.
Collapse
Affiliation(s)
- Yi-Bo Hou
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China
| | - Sheng Chang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China
| | - Song Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China
| | - Wei-Jie Zhang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China.
| |
Collapse
|
18
|
Schmitz R, Manook M, Fitch Z, Anwar I, DeLaura I, Olaso D, Choi A, Yoon J, Bae Y, Song M, Farris AB, Kwun J, Knechtle S. Belatacept and carfilzomib-based treatment for antibody-mediated rejection in a sensitized nonhuman primate kidney transplantation model. FRONTIERS IN TRANSPLANTATION 2023; 2:1230393. [PMID: 38993898 PMCID: PMC11235304 DOI: 10.3389/frtra.2023.1230393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 08/22/2023] [Indexed: 07/13/2024]
Abstract
Introduction One-third of HLA-incompatible kidney transplant recipients experience antibody mediated rejection (AMR) with limited treatment options. This study describes a novel treatment strategy for AMR consisting of proteasome inhibition and costimulation blockade with or without complement inhibition in a nonhuman primate model of kidney transplantation. Methods All rhesus macaques in the present study were sensitized to maximally MHC-mismatched donors by two sequential skin transplants prior to kidney transplant from the same donor. All primates received induction therapy with rhesus-specific ATG (rhATG) and were maintained on various immunosuppressive regimens. Primates were monitored postoperatively for signs of acute AMR, which was defined as worsening kidney function resistant to high dose steroid rescue therapy, and a rise in serum donor-specific antibody (DSA) levels. Kidney biopsies were performed to confirm AMR using Banff criteria. AMR treatment consisted of carfilzomib and belatacept for a maximum of four weeks with or without complement inhibitor. Results Treatment with carfilzomib and belatacept was well tolerated and no treatment-specific side effects were observed. After initiation of treatment, we observed a reduction of class I and class II DSA in all primates. Most importantly, primates had improved kidney function evident by reduced serum creatinine and BUN as well as increased urine output. A four-week treatment was able to extend graft survival by up to two months. Discussion In summary, combined carfilzomib and belatacept effectively treated AMR in our highly sensitized nonhuman primate model, resulting in normalization of renal function and prolonged allograft survival. This regimen may translate into clinical practice to improve outcomes of patients experiencing AMR.
Collapse
Affiliation(s)
- Robin Schmitz
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, United States
| | - Miriam Manook
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, United States
| | - Zachary Fitch
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, United States
| | - Imran Anwar
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, United States
| | - Isabel DeLaura
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, United States
| | - Danae Olaso
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, United States
| | - Ashley Choi
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, United States
| | - Janghoon Yoon
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, United States
| | - Yeeun Bae
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, United States
| | - Mingqing Song
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, United States
| | - Alton B. Farris
- Department of Pathology, Emory University School of Medicine, Atlanta, GA, United States
| | - Jean Kwun
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, United States
| | - Stuart Knechtle
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
19
|
Sasaki H, Tanabe T, Tsuji T, Hotta K. Mechanism and treatment for chronic antibody-mediated rejection in kidney transplant recipients. Int J Urol 2023; 30:624-633. [PMID: 37306194 DOI: 10.1111/iju.15197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 04/11/2023] [Indexed: 06/13/2023]
Abstract
Chronic antibody-mediated rejection of kidney transplantation is a major cause of late-stage graft loss. Donor-specific antibodies are the main cause of antibody-mediated rejection; in particular, de novo donor-specific antibodies are a risk factor for chronic active antibody-mediated rejection. The level of de novo donor-specific antibodies tends to increase with time throughout long-term graft survival. Donor-specific antibodies induce humoral rejection through complement activation, which results in tissue injury and coagulation. Additionally, complement activation promotes the migration of inflammatory cells through the innate immune response, causing endothelial injury. This inflammatory response may cause persistent glomerulitis and peritubular capillaritis, leading to fixed pathological lesions that impair graft function. No treatment has been established for chronic antibody-mediated rejection, a condition in which antibody-mediated rejection becomes irreversible. Thus, antibody-mediated rejection must be detected and treated while it is still reversible. In this review, we discuss the development of de novo donor-specific antibodies and the mechanisms leading to chronic antibody-mediated rejection and summarize the current treatment options and the latest biomarkers for detecting chronic antibody-mediated rejection at an earlier stage.
Collapse
Affiliation(s)
- Hajime Sasaki
- Division of Renal Surgery and Transplantation, Department of Urology, Jichi Medical University Hospital, Shimotsuke, Japan
- Department of Kidney Transplant Surgery, Sapporo City General Hospital, Sapporo, Japan
| | - Tatsu Tanabe
- Department of Kidney Transplant Surgery, Sapporo City General Hospital, Sapporo, Japan
| | - Takahiro Tsuji
- Department of Pathology, Sapporo City General Hospital, Sapporo, Japan
| | - Kiyohiko Hotta
- Department of Urology, Hokkaido University Hospital, Sapporo, Japan
| |
Collapse
|
20
|
Alasfar S, Kodali L, Schinstock CA. Current Therapies in Kidney Transplant Rejection. J Clin Med 2023; 12:4927. [PMID: 37568328 PMCID: PMC10419508 DOI: 10.3390/jcm12154927] [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: 06/19/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Despite significant advancements in immunosuppressive therapies, kidney transplant rejection continues to pose a substantial challenge, impacting the long-term survival of grafts. This article provides an overview of the diagnosis, current therapies, and management strategies for acute T-cell-mediated rejection (TCMR) and antibody-mediated rejection (ABMR). TCMR is diagnosed through histological examination of kidney biopsy samples, which reveal the infiltration of mononuclear cells into the allograft tissue. Corticosteroids serve as the primary treatment for TCMR, while severe or steroid-resistant cases may require T-cell-depleting agents, like Thymoglobulin. ABMR occurs due to the binding of antibodies to graft endothelial cells. The most common treatment for ABMR is plasmapheresis, although its efficacy is still a subject of debate. Other current therapies, such as intravenous immunoglobulins, anti-CD20 antibodies, complement inhibitors, and proteasome inhibitors, are also utilized to varying degrees, but their efficacy remains questionable. Management decisions for ABMR depend on the timing of the rejection episode and the presence of chronic changes. In managing both TCMR and ABMR, it is crucial to optimize immunosuppression and address adherence. However, further research is needed to explore newer therapeutics and evaluate their efficacy.
Collapse
Affiliation(s)
- Sami Alasfar
- Department of Medicine, Mayo Clinic, Phoenix, AZ 85054, USA;
| | - Lavanya Kodali
- Department of Medicine, Mayo Clinic, Phoenix, AZ 85054, USA;
| | | |
Collapse
|
21
|
Mizera J, Pilch J, Giordano U, Krajewska M, Banasik M. Therapy in the Course of Kidney Graft Rejection-Implications for the Cardiovascular System-A Systematic Review. Life (Basel) 2023; 13:1458. [PMID: 37511833 PMCID: PMC10381422 DOI: 10.3390/life13071458] [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: 06/01/2023] [Revised: 06/20/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Kidney graft failure is not a homogenous disease and the Banff classification distinguishes several types of graft rejection. The maintenance of a transplant and the treatment of its failure require specific medications and differ due to the underlying molecular mechanism. As a consequence, patients suffering from different rejection types will experience distinct side-effects upon therapy. The review is focused on comparing treatment regimens as well as presenting the latest insights into innovative therapeutic approaches in patients with an ongoing active ABMR, chronic active ABMR, chronic ABMR, acute TCMR, chronic active TCMR, borderline and mixed rejection. Furthermore, the profile of cardiovascular adverse effects in relation to the applied therapy was subjected to scrutiny. Lastly, a detailed assessment and comparison of different approaches were conducted in order to identify those that are the most and least detrimental for patients suffering from kidney graft failure.
Collapse
Affiliation(s)
- Jakub Mizera
- Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, 50-551 Wroclaw, Poland
| | - Justyna Pilch
- Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, 50-551 Wroclaw, Poland
| | - Ugo Giordano
- University Clinical Hospital, Wroclaw Medical University, 50-551 Wroclaw, Poland
| | - Magdalena Krajewska
- Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, 50-551 Wroclaw, Poland
| | - Mirosław Banasik
- Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, 50-551 Wroclaw, Poland
| |
Collapse
|
22
|
Parajuli S, Zhong W, Pantha M, Sokup M, Aziz F, Garg N, Mohamed M, Mandelbrot D. The Trend of Serum Creatinine Does Not Predict Follow-Up Biopsy Findings Among Kidney Transplant Recipients With Antibody-Mediated Rejection. Transplant Direct 2023; 9:e1489. [PMID: 37250486 PMCID: PMC10212615 DOI: 10.1097/txd.0000000000001489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 03/23/2023] [Indexed: 05/31/2023] Open
Abstract
Traditionally, antibody-mediated rejection (AMR) has been suspected mainly by a rise in serum creatinine (Scr) and confirmed by allograft biopsy. There is limited literature describing the trend of Scr after treatment, and how that trend might differ between patients with histological response and with no response to treatment. Methods We included all cases of AMR at our program between March 2016 and July 2020 who had a follow-up biopsy after the index biopsy, with initial diagnosis of AMR. We trended the Scr and change in Scr (delta Scr) and its association with being a responder (microvascular inflammation, MVI ≤1) or nonresponder (MVI >1), as well as graft failure. Results A total of 183 kidney transplant recipients were included, 66 in the responder group and 177 in the nonresponder group. The MVI scores and sum chronicity scores, along with transplant glomerulopathy scores, were higher in the nonresponder group. However, Scr at index biopsy was similar in responders (1.74 ± 0.70) versus nonresponders (1.83 ± 0.65; P = 0.39), as were the delta Scr at various time points. After adjustment for multiple variables, delta Scr was not associated with being a nonresponder. Also, delta Scr value at follow-up biopsy compared with index biopsy among responders was 0 ± 0.67 (P = 0.99) and among nonresponders was -0.01 ± 0.61 (P = 0.89). Being a nonresponder was significantly associated with an increased risk of graft failure at the last follow-up in univariate analysis but was not in multivariate analysis (hazard ratio 1.35; 95% confidence interval, 0.58-3.17; P = 0.49). Conclusions We found that Scr is not a good predictor of the resolution of MVI, supporting the utility of follow-up biopsies after treatment of AMR.
Collapse
Affiliation(s)
- Sandesh Parajuli
- Division of Nephrology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Weixiong Zhong
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Monika Pantha
- Division of Nephrology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Megan Sokup
- Division of Nephrology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Fahad Aziz
- Division of Nephrology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Neetika Garg
- Division of Nephrology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Maha Mohamed
- Division of Nephrology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Didier Mandelbrot
- Division of Nephrology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI
| |
Collapse
|
23
|
Stringer D, Gardner L, Shaw O, Clarke B, Briggs D, Worthington J, Buckland M, Danzi G, Hilton R, Picton M, Thuraisingham R, Borrows R, Baker R, McCullough K, Stoves J, Phanish M, Shah S, Shiu KY, Walsh SB, Ahmed A, Ayub W, Hegarty J, Tinch-Taylor R, Georgiou E, Bidad N, Kılıç A, Moon Z, Horne R, McCrone P, Kelly J, Murphy C, Peacock J, Dorling A. Optimized immunosuppression to prevent graft failure in renal transplant recipients with HLA antibodies (OuTSMART): a randomised controlled trial. EClinicalMedicine 2023; 56:101819. [PMID: 36684392 PMCID: PMC9852275 DOI: 10.1016/j.eclinm.2022.101819] [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: 09/30/2022] [Revised: 12/14/2022] [Accepted: 12/22/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND 3% of kidney transplant recipients return to dialysis annually upon allograft failure. Development of antibodies (Ab) against human leukocyte antigens (HLA) is a validated prognostic biomarker of allograft failure. We tested whether screening for HLA Ab, combined with an intervention to improve adherence and optimization of immunosuppression could prevent allograft failure. METHODS Prospective, open-labelled randomised biomarker-based strategy (hybrid) trial in 13 UK transplant centres [EudraCT (2012-004308-36) and ISRCTN (46157828)]. Patients were randomly allocated (1:1) to unblinded or double-blinded arms and screened every 8 months. Unblinded HLA Ab+ patients were interviewed to encourage medication adherence and had tailored optimisation of Tacrolimus, Mycophenolate mofetil and Prednisolone. The primary outcome was time to graft failure in an intention to treat analysis. The trial had 80% power to detect a hazard ratio of 0.49 in donor specific antibody (DSA)+ patients. FINDINGS From 11/9/13 to 27/10/16, 5519 were screened for eligibility and 2037 randomised (1028 to unblinded care and 1009 to double blinded care). We identified 198 with DSA and 818 with non-DSA. Development of DSA, but not non-DSA was predictive of graft failure. HRs for graft failure in unblinded DSA+ and non-DSA+ groups were 1.54 (95% CI: 0.72 to 3.30) and 0.97 (0.54-1.74) respectively, providing no evidence of an intervention effect. Non-inferiority for the overall unblinded versus blinded comparison was not demonstrated as the upper confidence limit of the HR for graft failure exceeded 1.4 (1.02, 95% CI: 0.72 to 1.44). The only secondary endpoint reduced in the unblinded arm was biopsy-proven rejection. INTERPRETATION Intervention to improve adherence and optimize immunosuppression does not delay failure of renal transplants after development of DSA. Whilst DSA predicts increased risk of allograft failure, novel interventions are needed before screening can be used to direct therapy. FUNDING The National Institute for Health Research Efficacy and Mechanism Evaluation programme grant (ref 11/100/34).
Collapse
Affiliation(s)
- Dominic Stringer
- Biostatistics and Health Informatics, The Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- King's Clinical Trials Unit, King's College London, London, UK
| | - Leanne Gardner
- King's Clinical Trials Unit, King's College London, London, UK
- Centre for Nephrology, Urology and Transplantation, Department of Inflammation Biology, King's College London, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Olivia Shaw
- Clinical Transplantation Laboratory, Viapath Analytics LLP, London, UK
| | - Brendan Clarke
- Transplant Immunology, Level 09 Gledhow Wing, St James's University Hospital, Beckett Street, Leeds, LS9 7TF, UK
| | - David Briggs
- NHSBT Birmingham, Vincent Drive, Edgbaston, Birmingham, B15 2SG, UK
| | - Judith Worthington
- Transplantation Laboratory, Manchester Royal Infirmary, Oxford Road, Manchester, M13 9WL, UK
| | - Matthew Buckland
- Clinical Transplantation Laboratory, The Royal London Hospital, 2nd Floor, Pathology and Pharmacy Building, 80 Newark Street, London, E1 1BB, UK
| | - Guilherme Danzi
- Renal Unit, Hospital das Clínicas da Universidade Federal de Pernambuco, Av. Prof. Moraes Rego, 1235 - Cidade Universitária, Recife - PE, 50670-901, Brazil
| | - Rachel Hilton
- Department of Nephrology and Transplantation, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Michael Picton
- Department of Renal Medicine, Manchester Royal Infirmary, Oxford Road, Manchester, M13 9WL, UK
| | - Raj Thuraisingham
- Department of Renal Medicine and Transplantation, Barts Health NHS Trust, London, E1 1BB, UK
| | - Richard Borrows
- Renal Unit, University Hospital Birmingham, Edgbaston, Birmingham, B15 2LN, UK
| | - Richard Baker
- Renal Unit, St James's University Hospital, Beckett Street, Leeds, LS9 7TF, UK
| | - Keith McCullough
- Renal Unit, York Teaching Hospital NHS Foundation Trust, York, YO31 8HE, UK
| | - John Stoves
- Renal Unit, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, BD5 0NA, UK
| | - Mysore Phanish
- Renal Unit, Epsom and St Helier University Hospitals NHS Trust, Surrey, UK
| | - Sapna Shah
- Renal Unit, King's College Hospital, London, SE5 9RJ, UK
| | - Kin Yee Shiu
- UCL Department of Renal Medicine, Royal Free London NHS Foundation Trust, London, NW3 2QG, UK
| | - Stephen B. Walsh
- UCL Department of Renal Medicine, Royal Free London NHS Foundation Trust, London, NW3 2QG, UK
| | - Aimun Ahmed
- Renal Unit, Lancashire Teaching Hospitals NHS Foundation Trust, Preston, PR2 9HT, UK
| | - Waqar Ayub
- Renal Unit, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK
| | - Janet Hegarty
- Renal Unit, Salford Royal NHS Foundation Trust, Salford, M6 8HD, UK
| | - Rose Tinch-Taylor
- Biostatistics and Health Informatics, The Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- King's Clinical Trials Unit, King's College London, London, UK
| | | | - Natalie Bidad
- Centre for Behavioural Medicine, UCL School of Pharmacy, University College London, London, WC1H 9JP, UK
| | - Ayşenur Kılıç
- Centre for Behavioural Medicine, UCL School of Pharmacy, University College London, London, WC1H 9JP, UK
| | - Zoe Moon
- Centre for Behavioural Medicine, UCL School of Pharmacy, University College London, London, WC1H 9JP, UK
| | - Robert Horne
- Centre for Behavioural Medicine, UCL School of Pharmacy, University College London, London, WC1H 9JP, UK
| | - Paul McCrone
- King's Clinical Trials Unit, King's College London, London, UK
- Faculty of Education, Health and Human Sciences, University of Greenwich, London, UK
| | - Joanna Kelly
- King's Clinical Trials Unit, King's College London, London, UK
| | - Caroline Murphy
- King's Clinical Trials Unit, King's College London, London, UK
| | - Janet Peacock
- School of Life Course and Population Sciences, King's College London, London, UK
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Dartmouth College, USA
| | - Anthony Dorling
- Centre for Nephrology, Urology and Transplantation, Department of Inflammation Biology, King's College London, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
- Corresponding author.
| |
Collapse
|
24
|
Sethi S, Jordan SC. Novel therapies for treatment of antibody-mediated rejection of the kidney. Curr Opin Organ Transplant 2023; 28:29-35. [PMID: 36579683 DOI: 10.1097/mot.0000000000001037] [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: 12/30/2022]
Abstract
PURPOSE OF REVIEW We aim to discuss current literature on novel therapies for antibody-mediated rejection (AMR) in kidney transplantation with a focus on chronic AMR. RECENT FINDINGS IL-6/IL-6 receptor blockers appear promising in the treatment of chronic AMR. Blocking this pathway was shown to reduce human leucocyte antigen-antibodies, improve histologic inflammation and increase T-regulatory cells. Based on experience in desensitization, IgG degrading endopeptidase, imlifidase, could be effective in AMR. There have been case reports describing the successful use of plasma cell/natural killer-cell-directed anti-CD38 antibody in the treatment of AMR. Off-target effects have been noted and strategies to mitigate these will be needed when using these agents. Complement inhibitors could be an effective add-on strategy to antibody-depleting therapies but their role in AMR needs to be better defined. Combining proteasome inhibitors and costimulation blockers has shown encouraging results in the prevention of AMR in animal models and is now being investigated in humans. Other novel strategies such as Fc neonatal receptor blockers which inhibit the recycling of pathogenic IgG and bispecific antibodies against B-cell maturation antigen/CD3+ T cells warrant further investigation. SUMMARY There are now a number of emerging therapies with varied targets and mechanism(s) of action that hold promise in the management of AMR and improving allograft survival.
Collapse
Affiliation(s)
- Supreet Sethi
- Division of Nephrology, Department of Medicine, Comprehensive Transplant Center, Cedars Sinai Medical Center, Los Angeles, California, USA
| | | |
Collapse
|
25
|
Anti-interleukin-6 Antibody Clazakizumab in Antibody-mediated Renal Allograft Rejection: Accumulation of Antibody-neutralized Interleukin-6 Without Signs of Proinflammatory Rebound Phenomena. Transplantation 2023; 107:495-503. [PMID: 35969004 DOI: 10.1097/tp.0000000000004285] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Blockade of interleukin-6 (IL-6) has emerged as a promising therapeutic option for antibody-mediated rejection. Subtherapeutic anti-IL-6 antibody level or treatment cessation following prolonged cytokine neutralization may result in proinflammatory rebound phenomena via accumulation of IL-6 and/or modulated gene expression of major components of the IL-6/IL-6 receptor (IL-6R) axis. METHODS We evaluated biologic material obtained from a randomized controlled, double-blind phase 2 trial designed to evaluate the safety and efficacy of the anti-IL-6 monoclonal antibody clazakizumab in late antibody-mediated rejection. Twenty kidney transplant recipients, allocated to clazakizumab or placebo, received 4-weekly doses over 12 wks, followed by a 40-wk extension where all recipients received clazakizumab. Serum proteins were detected using bead-based immunoassays and RNA transcripts using quantitative real-time polymerase chain reaction (peripheral blood) or microarray analysis (serial allograft biopsies). RESULTS Clazakizumab treatment resulted in a substantial increase in median total (bound and unbound to drug) serum IL-6 level (1.4, 8015, and 13 600 pg/mL at 0, 12, and 52 wks), but median level of free (unbound to drug) IL-6 did not increase (3.0, 2.3, and 2.3 pg/mL, respectively). Neutralization of IL-6 did not boost soluble IL-6R or leukocyte or allograft expression of IL-6, IL-6R, and glycoprotein 130 mRNA. Cessation of treatment at the end of the trial did not result in a meaningful increase in C-reactive protein or accelerated progression of graft dysfunction during 12 mo of follow-up. CONCLUSION Our results argue against clinically relevant rebound phenomena and modulation of major components of the IL-6/IL-6R axis following prolonged IL-6 neutralization with clazakizumab.
Collapse
|
26
|
Piñeiro GJ, Lazo-Rodriguez M, Ventura-Aguiar P, Ramirez-Bajo MJ, Banon-Maneus E, Lozano M, Cid J, Hierro-Garcia N, Cucchiari D, Revuelta I, Montagud-Marrahi E, Palou E, Bayés-Genís B, Campistol JM, Diekmann F, Rovira J. Extracorporeal Photopheresis Improves Graft Survival in a Full-Mismatch Rat Model of Kidney Transplantation. Transpl Int 2023; 36:10840. [PMID: 36713113 PMCID: PMC9876976 DOI: 10.3389/ti.2023.10840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 01/02/2023] [Indexed: 01/14/2023]
Abstract
Extracorporeal photopheresis (ECP) is an immunomodulatory therapy based on the infusion of autologous cellular products exposed to ultraviolet light (UV) in the presence of a photosensitizer. The study evaluates the ECP efficacy as induction therapy in a full-mismatch kidney transplant rat model. Dark Agouti to Lewis (DA-L) kidney transplant model has been established. ECP product was obtained from Lewis rat recipients after DA kidney graft transplantation (LewDA). Leukocytes of those LewDA rats were exposed to 8-methoxy psoralen, and illuminated with UV-A. The ECP doses assessed were 10 × 106 and 100 × 106 cells/time point. Lewis recipients received seven ECP infusions. DA-L model was characterized by the appearance of donor-specific antibodies (DSA) and kidney function deterioration from day three after kidney transplant. The dysfunction progressed rapidly until graft loss (6.1 ± 0.5 days). Tacrolimus at 0.25 mg/kg prolonged rat survival until 11.4 ± 0.7 days (p = 0.0004). In this context, the application of leukocytes from LewDA sensitized rats accelerated the rejection (8.7 ± 0.45, p = 0.0012), whereas ECP product at high dose extended kidney graft survival until 26.3 ± 7.3 days, reducing class I and II DSA in surviving rats. ECP treatment increases kidney graft survival in full-mismatch rat model of acute rejection and is a suitable immunomodulatory therapy to be explored in kidney transplantation.
Collapse
Affiliation(s)
- Gaston J. Piñeiro
- Department of Nephrology and Kidney Transplantation, Hospital Clinic de Barcelona, Barcelona, Spain,Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Marta Lazo-Rodriguez
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Pedro Ventura-Aguiar
- Department of Nephrology and Kidney Transplantation, Hospital Clinic de Barcelona, Barcelona, Spain,Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maria J. Ramirez-Bajo
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain,Red de Investigación Renal (REDINREN), Instituto de Salud Carlos III, Madrid, Spain
| | - Elisenda Banon-Maneus
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain,Red de Investigación Renal (REDINREN), Instituto de Salud Carlos III, Madrid, Spain
| | - Miquel Lozano
- Apheresis Unit, Department of Hemotherapy and Hemostasis, IDIBAPS, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Joan Cid
- Apheresis Unit, Department of Hemotherapy and Hemostasis, IDIBAPS, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Natalia Hierro-Garcia
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain,Red de Investigación Renal (REDINREN), Instituto de Salud Carlos III, Madrid, Spain
| | - David Cucchiari
- Department of Nephrology and Kidney Transplantation, Hospital Clinic de Barcelona, Barcelona, Spain,Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ignacio Revuelta
- Department of Nephrology and Kidney Transplantation, Hospital Clinic de Barcelona, Barcelona, Spain,Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain,Red de Investigación Renal (REDINREN), Instituto de Salud Carlos III, Madrid, Spain
| | - Enrique Montagud-Marrahi
- Department of Nephrology and Kidney Transplantation, Hospital Clinic de Barcelona, Barcelona, Spain,Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Eduard Palou
- Department of Immunology, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Beatriu Bayés-Genís
- Department of Nephrology and Kidney Transplantation, Hospital Clinic de Barcelona, Barcelona, Spain,Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Josep M. Campistol
- Department of Nephrology and Kidney Transplantation, Hospital Clinic de Barcelona, Barcelona, Spain,Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Fritz Diekmann
- Department of Nephrology and Kidney Transplantation, Hospital Clinic de Barcelona, Barcelona, Spain,Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain,Red de Investigación Renal (REDINREN), Instituto de Salud Carlos III, Madrid, Spain,*Correspondence: Fritz Diekmann, ; Jordi Rovira,
| | - Jordi Rovira
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain,*Correspondence: Fritz Diekmann, ; Jordi Rovira,
| |
Collapse
|
27
|
Zhu L, Guo Z, Zhao D, Sa R, Zhao G, Guo H, Chen G. Case report: Daratumumab for treatment of refractory late or chronic active antibody-mediated rejection in renal allograft recipients with high levels of de novo donor-specific antibodies. Front Immunol 2023; 13:1087597. [PMID: 36713391 PMCID: PMC9875042 DOI: 10.3389/fimmu.2022.1087597] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/02/2022] [Indexed: 01/13/2023] Open
Abstract
Background Late or chronic active antibody-mediated rejection (AMR) associated with de novo donor-specific antibodies (dnDSA) after renal transplantation is a great clinical challenge because it is often resistant to conventional therapies. Daratumumab, an anti-CD38 monoclonal antibody that can deplete plasma cells, may be effective for the treatment of late or chronic active AMR. Methods We designed a novel regimen that included early intensive therapy with daratumumab plus plasmapheresis (PP)/intravenous immunoglobulins (IVIG) and later maintenance therapy with daratumumab alone, and used this regimen to treat late or chronic active AMR in two kidney transplant recipients with extremely high levels of anti-DQ7 dnDSA. Results Both patients had a limited clinical response to the early treatment with rituximab and PP/IVIG (with or without splenic irradiation); however, they had a remarkable decrease in anti-DQ7 DSA (MFI value from ~20,000 to ~5,000) after 2-3 months of intensive therapy with daratumumab plus PP/IVIG. Over 20 months of follow-up, patient 1 maintained a low DSA (as low as 1,572) and normal renal function on daratumumab maintenance therapy. Patient 2 retained a low DSA and improved renal function and pathological lesions within one year after treatment but then deteriorated because of acute T cell-mediated rejection. Conclusions Our daratumumab-based regimen has shown promising results in the treatment of refractory late active or chronic active AMR in renal transplant recipients with high-level dnDSA. This may provide a reference for better use of daratumumab in the treatment of late or chronic active AMR.
Collapse
Affiliation(s)
- Lan Zhu
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China,Key Laboratory of Organ Transplantation, National Health Commission (NHC), Wuhan, China,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Zhiliang Guo
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Daqiang Zhao
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China,Key Laboratory of Organ Transplantation, National Health Commission (NHC), Wuhan, China,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Rula Sa
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guangyuan Zhao
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China,Key Laboratory of Organ Transplantation, National Health Commission (NHC), Wuhan, China,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Hui Guo
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China,Key Laboratory of Organ Transplantation, National Health Commission (NHC), Wuhan, China,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Gang Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China,Key Laboratory of Organ Transplantation, National Health Commission (NHC), Wuhan, China,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China,*Correspondence: Gang Chen,
| |
Collapse
|
28
|
Khairallah P, Robbins-Juarez S, Patel S, Shah V, Toma K, Fernandez H, Dube GK, King K, Mohan S, Husain SA, Morris H, Crew RJ. Tocilizumab for the treatment of chronic antibody mediated rejection in kidney transplant recipients. Clin Transplant 2023; 37:e14853. [PMID: 36398915 DOI: 10.1111/ctr.14853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 09/28/2022] [Accepted: 11/04/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Chronic active antibody-mediated rejection (CAAMR) constitutes a dominant form of late allograft failure. Several treatment strategies directed at CAAMR have been attempted but proven ineffective at delaying kidney function decline or reducing donor-specific antibodies (DSA). We describe our single-center experience using tocilizumab in patients with CAAMR. METHODS This is a retrospective analysis using electronic medical records. 38 kidney transplant recipients at Columbia University Irving Medical Center who had been prescribed tocilizumab and followed for at least 3 months between August 2013 through December 2019 were included. RESULTS Tocilizumab use was associated with a decrease in the rate of estimated glomerular filtration rate (eGFR) decline in the 6 months following treatment initiation as compared to the 3 months before tocilizumab was initiated (difference between slopes before and after initiation of treatment = 2.6 mL/min/1.73 m2 (SE = .8, p = .002) per month for up to 6 months following Tocilizumab initiation). Allograft biopsies showed significant improvement in interstitial inflammation scores (score 1(0,1) to 0 (0,1), p = .03) while other histologic scores remained stable. There was no significant change in proteinuria or DSA titers post-treatment with tocilizumab. CONCLUSIONS Treatment of CAAMR with tocilizumab was associated with a decrease in the rate of eGFR decline and a reduction in interstitial inflammation scores in patients with CAAMR.
Collapse
Affiliation(s)
| | - Shelief Robbins-Juarez
- Department of Internal Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Shefali Patel
- Division of Nephrology, Columbia University Irving Medical Center, New York, New York, USA
| | - Vaqar Shah
- Department of Medicine, SUNY University at Buffalo, Buffalo, New York, USA
| | - Katherine Toma
- Jersey Coast Nephrology and Hypertension Associates, Brick, New Jersey, USA
| | - Hilda Fernandez
- Division of Nephrology, Columbia University Irving Medical Center, New York, New York, USA
| | - Geoffrey K Dube
- Division of Nephrology, Columbia University Irving Medical Center, New York, New York, USA
| | - Kristen King
- Division of Nephrology, Columbia University Irving Medical Center, New York, New York, USA
| | - Sumit Mohan
- Division of Nephrology, Columbia University Irving Medical Center, New York, New York, USA
| | - Syed Ali Husain
- Division of Nephrology, Columbia University Irving Medical Center, New York, New York, USA
| | - Heather Morris
- Division of Nephrology, Columbia University Irving Medical Center, New York, New York, USA
| | - Russell John Crew
- Division of Nephrology, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
29
|
Tambur AR, Bestard O, Campbell P, Chong AS, Barrio MC, Ford ML, Gebel HM, Heidt S, Hickey M, Jackson A, Kosmoliaptsis V, Lefaucheur C, Louis K, Mannon RB, Mengel M, Morris A, Pinelli DF, Reed EF, Schinstock C, Taupin JL, Valenzuela N, Wiebe C, Nickerson P. Sensitization in transplantation: Assessment of Risk 2022 Working Group Meeting Report. Am J Transplant 2023; 23:133-149. [PMID: 36695615 DOI: 10.1016/j.ajt.2022.11.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/20/2022] [Accepted: 11/02/2022] [Indexed: 01/13/2023]
Abstract
The Sensitization in Transplantation: Assessment of Risk workgroup is a collaborative effort of the American Society of Transplantation and the American Society of Histocompatibility and Immunogenetics that aims at providing recommendations for clinical testing, highlights gaps in current knowledge, and proposes areas for further research to enhance histocompatibility testing in support of solid organ transplantation. This report provides updates on topics discussed by the previous Sensitization in Transplantation: Assessment of Risk working groups and introduces 2 areas of exploration: non-human leukocyte antigen antibodies and utilization of human leukocyte antigen antibody testing measurement to evaluate the efficacy of antibody-removal therapies.
Collapse
Affiliation(s)
- Anat R Tambur
- Department of Surgery, Comprehensive Transplant Center, Northwestern University, Chicago, Illinois, USA.
| | - Oriol Bestard
- Vall d'Hebron Institut de Recerca, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Patricia Campbell
- Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, Canada
| | - Anita S Chong
- Section of Transplantation, Department of Surgery, The University of Chicago, Chicago, Illinois, USA
| | - Martha Crespo Barrio
- Department of Nephrology, Hospital del Mar & Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Mandy L Ford
- Department of Surgery and Emory Transplant Center, Emory University, Atlanta, Georgia, USA
| | - Howard M Gebel
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sebastiaan Heidt
- Department of Immunology, Leiden University Medical Center, Netherlands
| | - Michelle Hickey
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Annette Jackson
- Department of Immunology, Duke University School of Medicine, Durham, North Carolina, USA
| | | | - Carmen Lefaucheur
- Paris Translational Research Center for Organ Transplantation, Institut national de la santé et de la recherche médicale UMR-S970, Université de Paris, Paris, France
| | - Kevin Louis
- Paris Translational Research Center for Organ Transplantation, Institut national de la santé et de la recherche médicale UMR-S970, Université de Paris, Paris, France
| | - Roslyn B Mannon
- Department of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Michael Mengel
- Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, Canada
| | - Anna Morris
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - David F Pinelli
- Department of Surgery, Comprehensive Transplant Center, Northwestern University, Chicago, Illinois, USA
| | - Elaine F Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | | | - Jean-Luc Taupin
- Department of Immunology, Saint Louis Hospital and University Paris-Cité, Paris, France
| | - Nicole Valenzuela
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Chris Wiebe
- Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Peter Nickerson
- Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
30
|
Clazakizumab for the treatment of chronic active antibody-mediated rejection (AMR) in kidney transplant recipients: Phase 3 IMAGINE study rationale and design. Trials 2022; 23:1042. [PMID: 36550562 PMCID: PMC9772593 DOI: 10.1186/s13063-022-06897-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 11/08/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Chronic active antibody-mediated rejection (AMR) is a major cause of graft loss with no approved drugs for its treatment. Currently, off-label regimens are used, reflecting the high unmet need for effective therapies based on well-controlled trials. Clazakizumab is a high-affinity, humanized monoclonal antibody that binds interleukin-6 and decreases donor-specific antibody (DSA) production and inflammation. Phase 2 pilot studies of clazakizumab in kidney transplant recipients with chronic active AMR suggest modulation of DSA, stabilization of glomerular filtration rate (GFR), and a manageable safety profile. We report the design of the Phase 3 IMAGINE study (NCT03744910) to evaluate the safety and efficacy of clazakizumab for the treatment of chronic active AMR. METHODS IMAGINE is a multicenter, double-blind trial of approximately 350 kidney transplant recipients with chronic active AMR (Banff chronic glomerulopathy [cg] >0 with concurrent positive human leukocyte antigen DSA) randomized 1:1 to receive clazakizumab or placebo (12.5 mg subcutaneous once every 4 weeks). The event-driven trial design will follow patients until 221 occurrences of all-cause graft loss are observed, defined as return to dialysis, graft nephrectomy, re-transplantation, estimated GFR (eGFR) <15 mL/min/1.73m2, or death from any cause. A surrogate for graft loss (eGFR slope) will be assessed at 1 year based on prior modeling validation. Secondary endpoints will include measures of pharmacokinetics/pharmacodynamics. Recruitment is ongoing across North America, Europe, Asia, and Australia. DISCUSSION IMAGINE represents the first Phase 3 clinical trial investigating the safety and efficacy of clazakizumab in kidney transplant recipients with chronic active AMR, and the largest placebo-controlled trial in this patient population. This trial includes prognostic biomarker enrichment and uniquely utilizes the eGFR slope at 1 year as a surrogate endpoint for graft loss, which may accelerate the approval of a novel therapy for patients at risk of graft loss. The findings of this study will be fundamental in helping to address the unmet need for novel therapies for chronic active AMR. TRIAL REGISTRATION ClinicalTrials.gov NCT03744910 . Registered on November 19, 2018.
Collapse
|
31
|
Nickerson PW. Rationale for the IMAGINE study for chronic active antibody-mediated rejection (caAMR) in kidney transplantation. Am J Transplant 2022; 22 Suppl 4:38-44. [PMID: 36453707 DOI: 10.1111/ajt.17210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 10/19/2022] [Indexed: 12/02/2022]
Abstract
Chronic active antibody-mediated rejection (caAMR) in kidney transplantation is a major cause of late graft loss and despite all efforts to date, there is no proven effective therapy. Indeed, the Transplant Society (TTS) consensus opinion called for a conservative approach optimizing baseline immunosuppression and supportive care focused on blood pressure, blood glucose, and lipid control. This review provides the rationale and early evidence in kidney transplant recipients with caAMR that supported the design of the IMAGINE study whose goal is to evaluate the potential impact of targeting the IL6/IL6R pathway.
Collapse
Affiliation(s)
- Peter W Nickerson
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada.,Department of Immunology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
32
|
Kervella D, Blancho G. New immunosuppressive agents in transplantation. Presse Med 2022; 51:104142. [PMID: 36252821 DOI: 10.1016/j.lpm.2022.104142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 09/29/2022] [Indexed: 11/24/2022] Open
Abstract
Immunosuppressive agents have enabled the development of allogenic transplantation during the last 40 years, allowing considerable improvement in graft survival. However, several issues remain such as the nephrotoxicity of calcineurin inhibitors, the cornerstone of immunosuppressive regimens and/or the higher risk of opportunistic infections and cancers. Most immunosuppressive agents target T cell activation and may not be efficient enough to prevent allo-immunization in the long term. Finally, antibody mediated rejection due to donor specific antibodies strongly affects allograft survival. Many drugs have been tested in the last decades, but very few have come to clinical use. The most recent one is CTLA4-Ig (belatacept), a costimulation blockade molecule that targets the second signal of T cell activation and is associated with a better long term kidney function than calcineurin inhibitors, despite an increased risk of acute cellular rejection. The research of new maintenance long-term immunosuppressive agents focuses on costimulation blockade. Agents inhibiting CD40-CD40 ligand interaction may enable a good control of both T cells and B cells responses. Anti-CD28 antibodies may promote regulatory T cells. Agents targeting this costimulation pathways are currently evaluated in clinical trials. Immunosuppressive agents for ABMR treatment are scarce since anti-CD20 agent rituximab and proteasome inhibitor bortezomib have failed to demonstrate an interest in ABMR. New drugs focusing on antibodies removal (imlifidase), B cell and plasmablasts (anti-IL-6/IL-6R, anti-CD38…) and complement inhibition are in the pipeline, with the challenge of their evaluation in such a heterogeneous pathology.
Collapse
Affiliation(s)
- Delphine Kervella
- CHU Nantes, Nantes Université, Service de Néphrologie et d'immunologie clinique, ITUN, Nantes, France; Nantes Université, CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, F-44000 Nantes, France
| | - Gilles Blancho
- CHU Nantes, Nantes Université, Service de Néphrologie et d'immunologie clinique, ITUN, Nantes, France; Nantes Université, CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, F-44000 Nantes, France.
| |
Collapse
|
33
|
van Vugt LK, Schagen MR, de Weerd A, Reinders ME, de Winter BC, Hesselink DA. Investigational drugs for the treatment of kidney transplant rejection. Expert Opin Investig Drugs 2022; 31:1087-1100. [PMID: 36175360 DOI: 10.1080/13543784.2022.2130751] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Kidney transplant rejection remains an important clinical problem despite the development of effective immunosuppressive drug combination therapy. Two major types of rejection are recognized, namely T-cell-mediated rejection (TCMR) and antibody-mediated rejection (ABMR), which have a different pathophysiology and are treated differently. Unfortunately, long-term outcomes of both TCMR and ABMR remain unsatisfactory despite current therapy. Hence, alternative therapeutic drugs are urgently needed. AREAS COVERED This review covers novel and investigational drugs for the pharmacological treatment of kidney transplant rejection. Potential therapeutic strategies and future directions are discussed. EXPERT OPINION The development of alternative pharmacologic treatment of rejection has focused mostly on ABMR, since this is the leading cause of kidney allograft loss and currently lacks an effective, evidence-based therapy. At present, there is insufficient high-quality evidence for any of the covered investigational drugs to support their use in ABMR. However, with the emergence of targeted therapies, this potential arises for individualized treatment strategies. In order to generate more high-quality evidence for such strategies and overcome the obstacles of classic, randomized, controlled trials, we advocate the implementation of adaptive trial designs and surrogate clinical endpoints. We believe such adaptive trial designs could help to understand the risks and benefits of promising drugs such as tocilizumab, clazakizumab, belimumab, and imlifidase.
Collapse
Affiliation(s)
- Lukas K van Vugt
- Erasmus MC Transplant Institute, Rotterdam, the Netherlands.,Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Maaike R Schagen
- Erasmus MC Transplant Institute, Rotterdam, the Netherlands.,Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Annelies de Weerd
- Erasmus MC Transplant Institute, Rotterdam, the Netherlands.,Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marlies Ej Reinders
- Erasmus MC Transplant Institute, Rotterdam, the Netherlands.,Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Brenda Cm de Winter
- Department of Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Dennis A Hesselink
- Erasmus MC Transplant Institute, Rotterdam, the Netherlands.,Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
34
|
Chamoun B, Sánchez-Sancho P, Torres IB, Gabaldon A, Perelló M, Sellarés J, Moreso F, Serón D. Tocilizumab in the treatment of active chronic humoral rejection resistant to standard therapy. Nefrologia 2022; 42:578-584. [PMID: 36717307 DOI: 10.1016/j.nefroe.2021.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 06/18/2023] Open
Abstract
INTRODUCTION There is no consensus on the most appropriate treatment for chronic active antibody-mediated rejection (cAMR). Recent studies suggest that treatment with tocilizumab (TCZ) may stabilize graft function, decrease the intensity of donor-specific HLA antibodies (DSAs) and reduce inflammation of microcirculation. PATIENTS AND METHODS Observational study with renal allograft recipients diagnosed with cAMR (n = 5) who had not submitted a response to traditional treatment based on the combination of plasma replacements, immunoglobulins, and rituximab. Patients were told to be treated with TCZ as compassionate use in six doses per month (8 mg/kg/month). Renal function, proteinuria, and the intensity of DSAs were monitored during follow-up. RESULTS Five patients, average age 60 ± 13 years, three male and two retrasplants (cPRA average 55%) with preformed DSAs. Treatment with TCZ was initiated within 47 ± 52 days of biopsy. In two cases treatment was discontinued after the first dose, by severe bicitopenia with cytomegalovirus viremia and by graft failure, respectively. In the three patients who completed treatment, no stability of renal function (serum creatinine from 1.73 ± 0.70 to 2.04 ± 0.52 mg/dL, e-FGR 4 6 ± 15 to 36 ± 16 mL/min), showed increased proteinuria (3.2 ± 4.0 to 6.9 ± 11.0 g/g) and the intensity of DSAs maintain stable. No changes were observed in the degree of inflammation of microcirculation (g+pt 4.2 ± 0.8 vs. 4.3 ± 1.0) or in the degree of transplant glomerulopathy (cg 1.2 ± 0.4 vs. 1.8 ± 1.0). CONCLUSIONS TCZ therapy does not appear to be effective in modifying the natural history of chronic active antibody-mediated rejection, does not improve the degree of inflammation of microcirculation and does not reduces the intensity of DSAs.
Collapse
Affiliation(s)
- Betty Chamoun
- Servicio de Nefrología, Hospital Universitari Vall d'Hebron, Universidad Autónoma de Barcelona, Barcelona, Spain.
| | - Pablo Sánchez-Sancho
- Servicio de Farmacia, Hospital Universitari Vall d'Hebron, Universidad Autónoma de Barcelona, Barcelona, Spain
| | - Irina B Torres
- Servicio de Nefrología, Hospital Universitari Vall d'Hebron, Universidad Autónoma de Barcelona, Barcelona, Spain.
| | - Alejandra Gabaldon
- Servicio de Patología, Hospital Universitari Vall d'Hebron, Universidad Autónoma de Barcelona, Barcelona, Spain
| | - Manel Perelló
- Servicio de Nefrología, Hospital Universitari Vall d'Hebron, Universidad Autónoma de Barcelona, Barcelona, Spain
| | - Joana Sellarés
- Servicio de Nefrología, Hospital Universitari Vall d'Hebron, Universidad Autónoma de Barcelona, Barcelona, Spain
| | - Francesc Moreso
- Servicio de Nefrología, Hospital Universitari Vall d'Hebron, Universidad Autónoma de Barcelona, Barcelona, Spain
| | - Daniel Serón
- Servicio de Nefrología, Hospital Universitari Vall d'Hebron, Universidad Autónoma de Barcelona, Barcelona, Spain
| |
Collapse
|
35
|
Chamoun B, Sánchez-Sancho P, Torres IB, Gabaldon A, Perelló M, Sellarés J, Moreso F, Serón D. Tocilizumab en el tratamiento del rechazo humoral crónico activo resistente a terapia estándar. Nefrologia 2022. [DOI: 10.1016/j.nefro.2021.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
36
|
Wei X, Xie Y, Jiang R, Li H, Wu H, Zhang Y, Li L, Zhou S, Ma X, Tang Z, He J, Wu D, Wu X. The impact of Rituximab administered before transplantation in patients undergoing allogeneic hematopoietic stem cell transplantation: A real-world study. Front Immunol 2022; 13:967026. [PMID: 36119024 PMCID: PMC9471377 DOI: 10.3389/fimmu.2022.967026] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Rituximab is used to eliminate B cells as a chimeric monoclonal antibody directed against CD20, a B-cell antigen expressed on B cells. To explore the impact of rituximab administered before transplantation, we implemented a retrospective, monocentric study and utilized real-world data collected at our center between January 2018 and December 2020, and then followed until December 2021. Based on whether a dose of 375mg/m2 rituximab was used at least once within two weeks before transplantation, patients undergoing allo-HSCT were classified into two groups: rituximab (N=176) and non-rituximab (N=344) group. Amongst all the patients, the application of rituximab decreased EBV reactivation (P<0.01) and rituximab was an independent factor in the prevention of EBV reactivation by both univariate and multivariate analyses (HR 0.56, 95%CI 0.33-0.97, P=0.04). In AML patients, there were significant differences in the cumulative incidence of aGVHD between the two groups (P=0.04). Our data showed that rituximab was association with a decreased incidence of aGVHD in AML patients according to both univariate and multivariate analyses. There was no difference between the two groups in other sets of populations. Thus, our study indicated that rituximab administered before transplantation may help prevent EBV reactivation in all allo-HSCT patients, as well as prevent aGVHD in AML patients after allo-HSCT.
Collapse
Affiliation(s)
- Xiya Wei
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yiyu Xie
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Internal Medicine, Yale-New Haven Health/Bridgeport Hospital, Bridgeport, CT, United States
| | - Ruoyu Jiang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Huiyu Li
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Heqing Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yuqi Zhang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Ling Li
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Shiyuan Zhou
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Xiao Ma
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Zaixiang Tang
- Department of Epidemiology and Statistics, School of Public Health, Faculty of Medicine, Soochow University, Suzhou, China
| | - Jun He
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- *Correspondence: Xiaojin Wu, ; Depei Wu,
| | - Xiaojin Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- *Correspondence: Xiaojin Wu, ; Depei Wu,
| |
Collapse
|
37
|
Yi SG, Gaber AO, Chen W. B-cell response in solid organ transplantation. Front Immunol 2022; 13:895157. [PMID: 36016958 PMCID: PMC9395675 DOI: 10.3389/fimmu.2022.895157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 07/11/2022] [Indexed: 11/21/2022] Open
Abstract
The transcriptional regulation of B-cell response to antigen stimulation is complex and involves an intricate network of dynamic signals from cytokines and transcription factors propagated from T-cell interaction. Long-term alloimmunity, in the setting of organ transplantation, is dependent on this B-cell response, which does not appear to be halted by current immunosuppressive regimens which are targeted at T cells. There is emerging evidence that shows that B cells have a diverse response to solid organ transplantation that extends beyond plasma cell antibody production. In this review, we discuss the mechanistic pathways of B-cell activation and differentiation as they relate to the transcriptional regulation of germinal center B cells, plasma cells, and memory B cells in the setting of solid organ transplantation.
Collapse
Affiliation(s)
- Stephanie G. Yi
- Division of Transplantation, Department of Surgery, Houston Methodist Hospital, Houston, TX, United States
- *Correspondence: Stephanie G. Yi,
| | - Ahmed Osama Gaber
- Division of Transplant Immunology, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States
| | - Wenhao Chen
- Division of Transplantation, Department of Surgery, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
38
|
Giannopoulou M, Tarassi K, Tsouka G, Christodoulidou C, Stefanidis I, Eleftheriadis T. Rituximab Administered for Recurrent Membranous Nephropathy in a Kidney Transplant Recipient Did Not Eliminate Donor-Specific Antibodies. EXP CLIN TRANSPLANT 2022; 20:695-697. [DOI: 10.6002/ect.2022.0146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
39
|
Mayer KA, Budde K, Jilma B, Doberer K, Böhmig GA. Emerging drugs for antibody-mediated rejection after kidney transplantation: a focus on phase II & III trials. Expert Opin Emerg Drugs 2022; 27:151-167. [PMID: 35715978 DOI: 10.1080/14728214.2022.2091131] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Antibody-mediated rejection (ABMR) is a leading cause of kidney allograft failure. Its therapy continues to be challenge, and no treatment has been approved for the market thus far. AREAS COVERED In this article, we discuss the pathophysiology and phenotypic presentation of ABMR, the current level of evidence to support the use of available therapeutic strategies, and the emergence of tailored drugs now being evaluated in systematic clinical trials. We searched PubMed, Clinicaltrials.gov and Citeline's Pharmaprojects for pertinent information on emerging anti-rejection strategies, laying a focus on phase II and III trials. EXPERT OPINION Currently, we rely on the use of apheresis for alloantibody depletion and intravenous immunoglobulin (referred to as standard of care), preferentially in early active ABMR. Recent systematic trials have questioned the benefits of using the CD20 antibody rituximab or the proteasome inhibitor bortezomib. However, there are now several promising treatment approaches in the pipeline, which are being trialed in phase II and III studies. These include interleukin-6 antagonism, CD38-targeting antibodies, and selective inhibitors of complement. On the basis of the information that has emerged so far, it seems that innovative treatment strategies for clinical use in ABMR may be available within the next 5-10 years.
Collapse
Affiliation(s)
- Katharina A Mayer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Klemens Budde
- Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Austria
| | - Konstantin Doberer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Georg A Böhmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
40
|
Naesens M, Budde K, Hilbrands L, Oberbauer R, Bellini MI, Glotz D, Grinyó J, Heemann U, Jochmans I, Pengel L, Reinders M, Schneeberger S, Loupy A. Surrogate Endpoints for Late Kidney Transplantation Failure. Transpl Int 2022; 35:10136. [PMID: 35669974 PMCID: PMC9163814 DOI: 10.3389/ti.2022.10136] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/18/2022] [Indexed: 12/13/2022]
Abstract
In kidney transplant recipients, late graft failure is often multifactorial. In addition, primary endpoints in kidney transplantation studies seek to demonstrate the short-term efficacy and safety of clinical interventions. Although such endpoints might demonstrate short-term improvement in specific aspects of graft function or incidence of rejection, such findings do not automatically translate into meaningful long-term graft survival benefits. Combining many factors into a well-validated model is therefore more likely to predict long-term outcome and better reflect the complexity of late graft failure than using single endpoints. If conditional marketing authorization could be considered for therapies that aim to improve long-term outcomes following kidney transplantation, then the surrogate endpoint for graft failure in clinical trial settings needs clearer definition. This Consensus Report considers the potential benefits and drawbacks of several candidate surrogate endpoints (including estimated glomerular filtration rate, proteinuria, histological lesions, and donor-specific anti-human leukocyte antigen antibodies) and composite scoring systems. The content was created from information prepared by a working group within the European Society for Organ Transplantation (ESOT). The group submitted a Broad Scientific Advice request to the European Medicines Agency (EMA), June 2020: the request focused on clinical trial design and endpoints in kidney transplantation. Following discussion and refinement, the EMA made final recommendations to ESOT in December 2020 regarding the potential to use surrogate endpoints in clinical studies that aim to improving late graft failure.
Collapse
Affiliation(s)
- Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Luuk Hilbrands
- Department of Nephrology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Rainer Oberbauer
- Department of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria
| | | | - Denis Glotz
- Paris Translational Research Center for Organ Transplantation, Hôpital Saint Louis, Paris, France
| | | | - Uwe Heemann
- Department of Nephrology, Technical University of Munich, Munich, Germany
| | - Ina Jochmans
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Liset Pengel
- Centre for Evidence in Transplantation, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Marlies Reinders
- Department of Internal Medicine, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Stefan Schneeberger
- Department of General, Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Alexandre Loupy
- Paris Translational Research Center for Organ Transplantation, Hôpital Necker, Paris, France
| |
Collapse
|
41
|
Wu K, Schmidt D, López del Moral C, Osmanodja B, Lachmann N, Halleck F, Choi M, Bachmann F, Ronicke S, Duettmann W, Naik M, Schrezenmeier E, Rudolph B, Budde K. Poor Outcomes in Patients With Transplant Glomerulopathy Independent of Banff Categorization or Therapeutic Interventions. Front Med (Lausanne) 2022; 9:889648. [PMID: 35646957 PMCID: PMC9133540 DOI: 10.3389/fmed.2022.889648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundTransplant glomerulopathy (TG) may indicate different disease entities including chronic AMR (antibody-mediated rejection). However, AMR criteria have been frequently changed, and long-term outcomes of allografts with AMR and TG according to Banff 2017 have rarely been investigated.Methods282 kidney allograft recipients with biopsy-proven TG were retrospectively investigated and diagnosed according to Banff'17 criteria: chronic AMR (cAMR, n = 72), chronic active AMR (cAAMR, n = 76) and isolated TG (iTG, n = 134). Of which 25/72 (34.7%) patients of cAMR group and 46/76 (60.5%) of cAAMR group were treated with antihumoral therapy (AHT).ResultsUp to 5 years after indication biopsy, no statistically significant differences were detected among iTG, cAMR and cAAMR groups in annual eGFR decline (−3.0 vs. −2.0 vs. −2.8 ml/min/1.73 m2 per year), 5-year median eGFR (21.5 vs. 16.0 vs. 20.0 ml/min/1.73 m2), 5-year graft survival rates (34.1 vs. 40.6 vs. 31.8%) as well as urinary protein excretion during follow-up. In addition, cAMR and cAAMR patients treated with AHT had similar graft and patient survival rates in comparison with those free of AHT, and similar comparing with iTG group. The TG scores were not associated with 5-year postbiopsy graft failure; whereas the patients with higher scores of chronic allograft scarring (by mm-, ci- and ct-lesions) had significantly lower graft survival rates than those with mild scores. The logistic-regression analysis demonstrated that Banff mm-, ah-, t-, ci-, ct-lesions and the eGFR level at biopsy were associated with 5-year graft failure.ConclusionsThe occurrence of TG is closely associated with graft failure independent of disease categories and TG score, and the long-term clinical outcomes were not influenced by AHT. The Banff lesions indicating progressive scarring might be better suited to predict an unfavorable outcome.
Collapse
Affiliation(s)
- Kaiyin Wu
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
- *Correspondence: Kaiyin Wu
| | - Danilo Schmidt
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
| | - Covadonga López del Moral
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
| | - Bilgin Osmanodja
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nils Lachmann
- HLA Laboratory, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| | - Fabian Halleck
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mira Choi
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
| | - Friederike Bachmann
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
| | - Simon Ronicke
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
| | - Wiebke Duettmann
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
| | - Marcel Naik
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
| | - Eva Schrezenmeier
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
| | - Birgit Rudolph
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universitätzu Berlin, Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
42
|
Borski A, Kainz A, Kozakowski N, Regele H, Kläger J, Strassl R, Fischer G, Faé I, Wenda S, Kikić Ž, Bond G, Reindl-Schwaighofer R, Mayer KA, Eder M, Wahrmann M, Haindl S, Doberer K, Böhmig GA, Eskandary F. Early Estimated Glomerular Filtration Rate Trajectories After Kidney Transplant Biopsy as a Surrogate Endpoint for Graft Survival in Late Antibody-Mediated Rejection. Front Med (Lausanne) 2022; 9:817127. [PMID: 35530045 PMCID: PMC9069161 DOI: 10.3389/fmed.2022.817127] [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: 11/17/2021] [Accepted: 03/14/2022] [Indexed: 11/25/2022] Open
Abstract
Background Late antibody-mediated rejection (ABMR) after kidney transplantation is a major cause of long-term allograft loss with currently no proven treatment strategy. Design for trials testing treatment for late ABMR poses a major challenge as hard clinical endpoints require large sample sizes. We performed a retrospective cohort study applying commonly used selection criteria to evaluate the slope of the estimated glomerular filtration rate (eGFR) within an early and short timeframe after biopsy as a surrogate of future allograft loss for clinical trials addressing late ABMR. Methods Study subjects were identified upon screening of the Vienna transplant biopsy database. Main inclusion criteria were (i) a solitary kidney transplant between 2000 and 2013, (ii) diagnosis of ABMR according to the Banff 2015 scheme at >12 months post-transplantation, (iii) age 15-75 years at ABMR diagnosis, (iv) an eGFR > 25 mL/min/1.73 m2 at ABMR diagnosis, and (v) a follow-up for at least 36 months after ABMR diagnosis. The primary outcome variable was death-censored graft survival. A mixed effects model with linear splines was used for eGFR slope modeling and association of graft failure and eGFR slope was assessed applying a multivariate competing risk analysis with landmarks set at 12 and 24 months after index biopsy. Results A total of 70 allografts from 68 patients were included. An eGFR loss of 1 ml/min/1.73 m2 per year significantly increased the risk for allograft failure, when eGFR slopes were modeled over 12 months [HR 1.1 (95% CI: 1.01-1.3), p = 0.020] or over 24 months [HR 1.3 (95% CI: 1.1-1.4), p = 0.001] after diagnosis of ABMR with landmarks set at both time points. Covariables influencing graft loss in all models were histologic evidence of glomerulonephritis concurring with ABMR as well as the administration of anti-thymocyte globulin (ATG) at the time of transplantation. Conclusion Our study supports the use of the eGFR slope modeled for at least 12 months after biopsy-proven diagnosis of late ABMR, as a surrogate parameter for future allograft loss. The simultaneous occurrence of glomerulonephritis together with ABMR at index biopsy and the use of ATG at the time of transplantation-likely representing a confounder in pre-sensitized recipients-were strongly associated with worse transplant outcomes.
Collapse
Affiliation(s)
- Anita Borski
- Department of Nephrology and Dialysis, Medical University Vienna, Vienna, Austria
| | - Alexander Kainz
- Department of Nephrology and Dialysis, Medical University Vienna, Vienna, Austria
| | | | - Heinz Regele
- Department of Pathology, Medical University Vienna, Vienna, Austria
| | - Johannes Kläger
- Department of Pathology, Medical University Vienna, Vienna, Austria
| | - Robert Strassl
- Division of Clinical Virology, Department of Laboratory Medicine, Medical University Vienna, Vienna, Austria
| | - Gottfried Fischer
- Department of Blood Group Serology and Transfusion Medicine, Medical University Vienna, Vienna, Austria
| | - Ingrid Faé
- Department of Blood Group Serology and Transfusion Medicine, Medical University Vienna, Vienna, Austria
| | - Sabine Wenda
- Department of Blood Group Serology and Transfusion Medicine, Medical University Vienna, Vienna, Austria
| | - Željko Kikić
- Department of Urology, Medical University Vienna, Vienna, Austria
| | - Gregor Bond
- Department of Nephrology and Dialysis, Medical University Vienna, Vienna, Austria
| | | | - Katharina A. Mayer
- Department of Nephrology and Dialysis, Medical University Vienna, Vienna, Austria
| | - Michael Eder
- Department of Nephrology and Dialysis, Medical University Vienna, Vienna, Austria
| | - Markus Wahrmann
- Department of Nephrology and Dialysis, Medical University Vienna, Vienna, Austria
| | - Susanne Haindl
- Department of Nephrology and Dialysis, Medical University Vienna, Vienna, Austria
| | - Konstantin Doberer
- Department of Nephrology and Dialysis, Medical University Vienna, Vienna, Austria
| | - Georg A. Böhmig
- Department of Nephrology and Dialysis, Medical University Vienna, Vienna, Austria
| | - Farsad Eskandary
- Department of Nephrology and Dialysis, Medical University Vienna, Vienna, Austria
| |
Collapse
|
43
|
Mayer KA, Budde K, Halloran PF, Doberer K, Rostaing L, Eskandary F, Christamentl A, Wahrmann M, Regele H, Schranz S, Ely S, Firbas C, Schörgenhofer C, Kainz A, Loupy A, Härtle S, Boxhammer R, Jilma B, Böhmig GA. Safety, tolerability, and efficacy of monoclonal CD38 antibody felzartamab in late antibody-mediated renal allograft rejection: study protocol for a phase 2 trial. Trials 2022; 23:270. [PMID: 35395951 PMCID: PMC8990453 DOI: 10.1186/s13063-022-06198-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 03/25/2022] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Antibody-mediated rejection (ABMR) is a cardinal cause of renal allograft loss. This rejection type, which may occur at any time after transplantation, commonly presents as a continuum of microvascular inflammation (MVI) culminating in chronic tissue injury. While the clinical relevance of ABMR is well recognized, its treatment, particularly a long time after transplantation, has remained a big challenge. A promising strategy to counteract ABMR may be the use of CD38-directed treatment to deplete alloantibody-producing plasma cells (PC) and natural killer (NK) cells. METHODS This investigator-initiated trial is planned as a randomized, placebo-controlled, double-blind, parallel-group, multi-center phase 2 trial designed to assess the safety and tolerability (primary endpoint), pharmacokinetics, immunogenicity, and efficacy of the fully human CD38 monoclonal antibody felzartamab (MOR202) in late ABMR. The trial will include 20 anti-HLA donor-specific antibody (DSA)-positive renal allograft recipients diagnosed with active or chronic active ABMR ≥ 180 days post-transplantation. Subjects will be randomized 1:1 to receive felzartamab (16 mg/kg per infusion) or placebo for a period of 6 months (intravenous administration on day 0, and after 1, 2, 3, 4, 8, 12, 16, and 20 weeks). Two follow-up allograft biopsies will be performed at weeks 24 and 52. Secondary endpoints (preliminary assessment) will include morphologic and molecular rejection activity in renal biopsies, immunologic biomarkers in the blood and urine, and surrogate parameters predicting the progression to allograft failure (slope of renal function; iBOX prediction score). DISCUSSION Based on the hypothesis that felzartamab is able to halt the progression of ABMR via targeting antibody-producing PC and NK cells, we believe that our trial could potentially provide the first proof of concept of a new treatment in ABMR based on a prospective randomized clinical trial. TRIAL REGISTRATION EU Clinical Trials Register (EudraCT) 2021-000545-40 . Registered on 23 June 2021. CLINICALTRIALS gov NCT05021484 . Registered on 25 August 2021.
Collapse
Affiliation(s)
- Katharina A Mayer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Währinger Gürtel 18-20, A-1090, Vienna, Austria
| | - Klemens Budde
- Department of Nephrology, Charité University Medicine Berlin, Berlin, Germany
| | - Philip F Halloran
- Alberta Transplant Applied Genomics Centre, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Konstantin Doberer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Währinger Gürtel 18-20, A-1090, Vienna, Austria
| | - Lionel Rostaing
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, University Hospital Grenoble, Grenoble, France
| | - Farsad Eskandary
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Währinger Gürtel 18-20, A-1090, Vienna, Austria
| | - Anna Christamentl
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Währinger Gürtel 18-20, A-1090, Vienna, Austria
| | - Markus Wahrmann
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Währinger Gürtel 18-20, A-1090, Vienna, Austria
| | - Heinz Regele
- Department of Clinical Pathology, Medical University of Vienna, Währinger Gürtel 18-20, A-1090, Vienna, Austria
| | - Sabine Schranz
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Sarah Ely
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Christa Firbas
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | | | - Alexander Kainz
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Währinger Gürtel 18-20, A-1090, Vienna, Austria
| | - Alexandre Loupy
- INSERM UMR 970, Paris Translational Research Centre for Organ Transplantation, Université de Paris, Paris, France
| | | | | | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria.
| | - Georg A Böhmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Währinger Gürtel 18-20, A-1090, Vienna, Austria.
| |
Collapse
|
44
|
Kong F, Ye S, Zhong Z, Zhou X, Zhou W, Liu Z, Lan J, Xiong Y, Ye Q. Single-Cell Transcriptome Analysis of Chronic Antibody-Mediated Rejection After Renal Transplantation. Front Immunol 2022; 12:767618. [PMID: 35111153 PMCID: PMC8801944 DOI: 10.3389/fimmu.2021.767618] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 12/27/2021] [Indexed: 12/13/2022] Open
Abstract
Renal transplantation is currently the most effective treatment for end-stage renal disease. However, chronic antibody-mediated rejection (cABMR) remains a serious obstacle for the long-term survival of patients with renal transplantation and a problem to be solved. At present, the role and mechanism underlying immune factors such as T- and B- cell subsets in cABMR after renal transplantation remain unclear. In this study, single-cell RNA sequencing (scRNA-seq) of peripheral blood monocytes (PBMCs) from cABMR and control subjects was performed to define the transcriptomic landscape at single-cell resolution. A comprehensive scRNA-seq analysis was performed. The results indicated that most cell types in the cABMR patients exhibited an intense interferon response and release of proinflammatory cytokines. In addition, we found that the expression of MT-ND6, CXCL8, NFKBIA, NFKBIZ, and other genes were up-regulated in T- and B-cells and these genes were associated with pro-inflammatory response and immune regulation. Western blot and qRT-PCR experiments also confirmed the up-regulated expression of these genes in cABMR. GO and KEGG enrichment analyses indicated that the overexpressed genes in T- and B-cells were mainly enriched in inflammatory pathways, including the TNF, IL-17, and Toll-like receptor signaling pathways. Additionally, MAPK and NF-κB signaling pathways were also involved in the occurrence and development of cABMR. This is consistent with the experimental results of Western blot. Trajectory analysis assembled the T-cell subsets into three differentiation paths with distinctive phenotypic and functional prog rams. CD8 effector T cells and γδ T cells showed three different differentiation trajectories, while CD8_MAI T cells and naive T cells primarily had two differentiation trajectories. Cell-cell interaction analysis revealed strong T/B cells and neutrophils activation in cABMR. Thus, the study offers new insight into pathogenesis and may have implications for the identification of novel therapeutic targets for cABMR.
Collapse
Affiliation(s)
- Fanhua Kong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, China.,National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Wuhan, China
| | - Shaojun Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, China.,National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Wuhan, China
| | - Zibiao Zhong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, China.,National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Wuhan, China
| | - Xin Zhou
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, China.,National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Wuhan, China
| | - Wei Zhou
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, China.,National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Wuhan, China
| | - Zhongzhong Liu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, China.,National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Wuhan, China
| | - Jianan Lan
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, China.,National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Wuhan, China
| | - Yan Xiong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, China.,National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Wuhan, China
| | - Qifa Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, China.,National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Wuhan, China.,The 3rd Xiangya Hospital of Central South University, Research Center of National Health Ministry on Transplantation Medicine Engineering and Technology, Changsha, China
| |
Collapse
|
45
|
Bertacchi M, Parvex P, Villard J. Antibody-mediated rejection after kidney transplantation in children; therapy challenges and future potential treatments. Clin Transplant 2022; 36:e14608. [PMID: 35137982 PMCID: PMC9286805 DOI: 10.1111/ctr.14608] [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: 11/09/2021] [Revised: 01/14/2022] [Accepted: 01/31/2022] [Indexed: 11/27/2022]
Abstract
Antibody‐mediated rejection (AMR) remains one of the most critical problems in renal transplantation, with a significant impact on patient and graft survival. In the United States, no treatment has received FDA approval jet. Studies about treatments of AMR remain controversial, limited by the absence of a gold standard and the difficulty in creating large, multi‐center studies. These limitations emerge even more in pediatric transplantation because of the limited number of pediatric studies and the occasional use of some therapies with unknown and poorly documented side effects. The lack of recommendations and the unsharp definition of different forms of AMR contribute to the challenging management of the therapy by pediatric nephrologists. In an attempt to help clinicians involved in the care of renal transplanted children affected by an AMR, we rely on the latest recommendations of the Transplantation Society (TTS) for the classification and treatment of AMR to describe treatments available today and potential new treatments with a particular focus on the pediatric population.
Collapse
Affiliation(s)
| | - Paloma Parvex
- Division of Pediatric Nephrology, University Children Hospital of Geneva, Geneva, Switzerland
| | - Jean Villard
- Division of Nephrology, University Hospital of Geneva, Geneva, Switzerland.,Division of Transplantation Immunology, University Hospital of Geneva, Geneva, Switzerland
| |
Collapse
|
46
|
Tanaka R, Tsutahara K, Inoguchi S, Horitani H, Asakura T, Kawamura N, Kakuta Y, Nakagawa M, Takao T. Clinical effect of rabbit anti-thymocyte globulin for chronic active antibody-mediated rejection after kidney transplantation. CEN Case Rep 2022; 11:79-83. [PMID: 34374932 PMCID: PMC8811014 DOI: 10.1007/s13730-021-00633-7] [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: 10/13/2020] [Accepted: 08/01/2021] [Indexed: 11/29/2022] Open
Abstract
Chronic active antibody-mediated rejection (CAAMR) is a frequent cause of late graft loss. However, effective treatment for CAAMR after kidney transplantation has not yet been established. Here, we present the case of a kidney transplant recipient who recovered from CAAMR after administration of rabbit anti-thymocyte globulin. A 61-year-old man underwent ABO-compatible living-donor kidney transplantation for end-stage kidney disease; the kidney was donated by his wife. Five years after the transplant, the patient's serum creatinine level and urine protein-to-creatinine ratio increased. He was subsequently diagnosed with CAAMR based on the kidney allograft biopsy and the presence of donor-specific human leukocyte antigen antibodies. Rabbit anti-thymocyte globulin treatment was administered following steroid pulse therapy. Subsequently, his serum creatinine levels and urine protein to creatinine ratio improved. There was also an improvement in the pathological findings seen on biopsy and the mean fluorescence intensity of donor-specific antibodies. In conclusion, this report describes the case of a kidney transplant recipient who developed CAAMR, treated using rabbit anti-thymocyte globulin. This strategy might be a viable treatment option for CAAMR after a kidney transplant.
Collapse
Affiliation(s)
- Ryo Tanaka
- Department of Urology, Osaka General Medical Center, 3-1-56 Bandaihigashi, Shumiyoshiku, Osaka City, Osaka, 558-8558, Japan
| | - Koichi Tsutahara
- Department of Urology, Osaka General Medical Center, 3-1-56 Bandaihigashi, Shumiyoshiku, Osaka City, Osaka, 558-8558, Japan.
| | - Shunsuke Inoguchi
- Department of Urology, Osaka General Medical Center, 3-1-56 Bandaihigashi, Shumiyoshiku, Osaka City, Osaka, 558-8558, Japan
| | - Hiromu Horitani
- Department of Urology, Osaka General Medical Center, 3-1-56 Bandaihigashi, Shumiyoshiku, Osaka City, Osaka, 558-8558, Japan
| | - Toshihisa Asakura
- Department of Urology, Osaka General Medical Center, 3-1-56 Bandaihigashi, Shumiyoshiku, Osaka City, Osaka, 558-8558, Japan
| | - Norihiko Kawamura
- Department of Urology, Osaka General Medical Center, 3-1-56 Bandaihigashi, Shumiyoshiku, Osaka City, Osaka, 558-8558, Japan
| | - Yoichi Kakuta
- Department of Urology, Osaka General Medical Center, 3-1-56 Bandaihigashi, Shumiyoshiku, Osaka City, Osaka, 558-8558, Japan
| | - Masahiro Nakagawa
- Department of Urology, Osaka General Medical Center, 3-1-56 Bandaihigashi, Shumiyoshiku, Osaka City, Osaka, 558-8558, Japan
| | - Tetsuya Takao
- Department of Urology, Osaka General Medical Center, 3-1-56 Bandaihigashi, Shumiyoshiku, Osaka City, Osaka, 558-8558, Japan
| |
Collapse
|
47
|
Koslik MA, Friebus-Kardash J, Heinemann FM, Kribben A, Bräsen JH, Eisenberger U. Differential Treatment Effects for Renal Transplant Recipients With DSA-Positive or DSA-Negative Antibody-Mediated Rejection. Front Med (Lausanne) 2022; 9:816555. [PMID: 35174191 PMCID: PMC8841765 DOI: 10.3389/fmed.2022.816555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/07/2022] [Indexed: 12/22/2022] Open
Abstract
Background Antibody-mediated rejection (ABMR) is the main cause of renal allograft loss. The most common treatment strategy is based on plasmapheresis plus the subsequent administration of intravenous immunoglobulin (IVIG). Unfortunately, no approved long-term therapy is available for ABMR. The current study was designed to analyze the effect of various ABMR treatment approaches on allograft survival and to compare treatment effects in the presence or absence of donor-specific antibodies (DSAs). Methods This single-center study retrospectively analyzed 102 renal allograft recipients who had biopsy-proven ABMR after transplant. DSA was detectable in 61 of the 102 patients. Initial standard treatment of ABMR consisted of plasmapheresis (PS) or immunoadsorption (IA), followed by a single course of IVIG. In case of nonresponse or recurrence, additional immunosuppressive medications, such as rituximab, bortezomib, thymoglobulin, or eculizumab, were administered. In a second step, persistent ABMR was treated with increased maintenance immunosuppression, long-term therapy with IVIG (more than 1 year), or both. Results Overall graft survival among transplant patients with ABMR was <50% after 3 years of follow-up. Compared to the use of PS/IA and IVIG alone, the use of additional immunosuppressive medications had no beneficial effect on allograft survival (p = 0.83). Remarkably, allografts survival rates were comparable between patients treated with the combination of PS/IA and IVIG and those treated with a single administration of IVIG (p = 0.18). Renal transplant patients with ABMR but without DSAs benefited more from increased maintenance immunosuppression than did DSA-positive patients with ABMR (p = 0.01). Recipients with DSA-positive ABMR exhibited significantly better allograft survival after long-term application of IVIG for more than 1 year than did recipients with DSA-negative ABMR (p = 0.02). Conclusions The results of our single-center cohort study involving kidney transplant recipients with ABMR suggest that long-term application of IVIG is more favorable for DSA-positive recipients, whereas intensification of maintenance immunosuppression is more effective for recipients with DSA-negative ABMR.
Collapse
Affiliation(s)
- Marius Andreas Koslik
- Department of Nephrology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Justa Friebus-Kardash
- Department of Nephrology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Falko Markus Heinemann
- Institute for Transfusion Medicine, Transplantation Diagnostics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Andreas Kribben
- Department of Nephrology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jan Hinrich Bräsen
- Nephropathology Unit, Hannover Medical School, Institute of Pathology, Hanover, Germany
| | - Ute Eisenberger
- Department of Nephrology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- *Correspondence: Ute Eisenberger
| |
Collapse
|
48
|
Noble J, Giovannini D, Laamech R, Imerzoukene F, Janbon B, Marchesi L, Malvezzi P, Jouve T, Rostaing L. Tocilizumab in the Treatment of Chronic Antibody-Mediated Rejection Post Kidney Transplantation: Clinical and Histological Monitoring. Front Med (Lausanne) 2022; 8:790547. [PMID: 35004757 PMCID: PMC8739887 DOI: 10.3389/fmed.2021.790547] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction: Chronic antibody-mediated rejection (cAMR) has very few effective therapeutic options. Interleukin-6 is an attractive target because it is involved in inflammation and humoral immunity. Therefore, the use of tocilizumab (anti-IL6 receptor, TCZ) is a potential valuable therapeutic option to treat cABMR in kidney-transplant (KT) recipients. Materials and Methods: This single-center retrospective study included all KT recipients that received monthly TCZ infusions in the setting of cABMR, between August 2018 and July 2021. We assessed 12-month renal function and KT histology during follow-up. Results: Forty patients were included. At 12-months, eGFR was not significantly different, 41.6 ± 17 vs. 43 ± 17 mL/min/1.73 m2 (p = 0.102) in patients with functional graft. Six patients (15%) lost their graft: their condition was clinically more severe at the time of first TCZ infusion. Histological follow-up showed no statistical difference in the scores of glomerulitis, peritubular capillaritis, and interstitial fibrosis/tubular atrophy (IFTA). Chronic glomerulopathy score however, increased significantly over time; conversely arteritis and inflammation in IFTA ares improved in follow-up biopsies. Conclusion: In our study, the addition of TCZ prevented clinical and histological worsening of cABMR in KT recipients, except for more severely ill patients. Randomized studies are needed to clarify the risk/benefit of TCZ in cABMR.
Collapse
Affiliation(s)
- Johan Noble
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, University Hospital Grenoble, Grenoble, France.,University Grenoble Alpes, Grenoble, France
| | - Diane Giovannini
- Pathology Department, University Hospital Grenoble, Grenoble, France
| | - Reda Laamech
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, University Hospital Grenoble, Grenoble, France
| | - Farida Imerzoukene
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, University Hospital Grenoble, Grenoble, France
| | - Bénédicte Janbon
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, University Hospital Grenoble, Grenoble, France
| | - Laura Marchesi
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, University Hospital Grenoble, Grenoble, France
| | - Paolo Malvezzi
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, University Hospital Grenoble, Grenoble, France
| | - Thomas Jouve
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, University Hospital Grenoble, Grenoble, France.,University Grenoble Alpes, Grenoble, France
| | - Lionel Rostaing
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, University Hospital Grenoble, Grenoble, France.,University Grenoble Alpes, Grenoble, France
| |
Collapse
|
49
|
Lasa-Lázaro M, Ramos-Boluda E, Mancebo E, Castro-Panete MJ, González-Sacristán R, Serradilla J, Andrés-Moreno AM, Hernández-Oliveros F, Paz-Artal E, Talayero P. Antibody-removal therapies for de novo DSA in pediatric intestinal recipients: Why, when, and how? A single-center experience. Front Pediatr 2022; 10:1074577. [PMID: 36819192 PMCID: PMC9932897 DOI: 10.3389/fped.2022.1074577] [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: 10/19/2022] [Accepted: 12/21/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Donor-specific anti-HLA antibodies (DSA) impact negatively on the outcome of intestinal grafts. Although the use of antibody-removal therapies (ART) is becoming more frequent in the last few years, issues regarding their timing and effectiveness remain under discussion. METHODS In the present study, we report our experience with eight ART procedures (based on plasmapheresis, intravenous immunoglobulin, and rituximab) in eight pediatric intestinal and multivisceral transplants with de novo DSA (dnDSA). RESULTS ART were performed when dnDSA appeared in two contexts: (1) concomitant with rejection (acute or chronic) or (2) without rejection or any other clinical symptom. Complete DSA removal was observed in seven out of eight patients, showing an effectiveness of 88%. In the group treated for dnDSA without clinical symptoms, the success rate was 100%, with complete DSA removal and without rejection afterward. A shorter time between DSA detection and ART performance appeared as a significant factor for the success of the therapy (p = 0.0002). DSA against HLA-A and DQ alleles were the most resistant to ART, whereas anti-DR DSA were the most sensitive. In addition, the 8-year allograft survival rate in recipients undergoing ART was similar to that in those without DSA, being significantly lower in non-treated DSA-positive recipients (p = 0.013). CONCLUSION The results confirm the effectiveness of ART in terms of DSA removal and allograft survival and encourage its early use even in the absence of clinical symptoms.
Collapse
Affiliation(s)
- María Lasa-Lázaro
- Department of Immunology, University Hospital 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Esther Ramos-Boluda
- Unit of Intestinal Rehabilitation and Transplant, University Hospital La Paz, Madrid, Spain
| | - Esther Mancebo
- Department of Immunology, University Hospital 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - María José Castro-Panete
- Department of Immunology, University Hospital 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | | | - Javier Serradilla
- Department of Pediatric Surgery, University Hospital La Paz, Madrid, Spain.,IdiPaz Research Institute, University Hospital La Paz, Madrid, Spain
| | - Ane Miren Andrés-Moreno
- Department of Pediatric Surgery, University Hospital La Paz, Madrid, Spain.,IdiPaz Research Institute, University Hospital La Paz, Madrid, Spain
| | - Francisco Hernández-Oliveros
- Department of Pediatric Surgery, University Hospital La Paz, Madrid, Spain.,IdiPaz Research Institute, University Hospital La Paz, Madrid, Spain
| | - Estela Paz-Artal
- Department of Immunology, University Hospital 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain.,School of Medicine, Complutense University, Madrid, Spain.,CIBER de Enfermedades Infecciosas, ISCIII, Madrid, Spain
| | - Paloma Talayero
- Department of Immunology, University Hospital 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| |
Collapse
|
50
|
Wu K, Schmidt D, López del Moral C, Osmanodja B, Lachmann N, Zhang Q, Halleck F, Choi M, Bachmann F, Ronicke S, Duettmann W, Naik MG, Schrezenmeier E, Rudolph B, Budde K. Poor Long-Term Renal Allograft Survival in Patients with Chronic Antibody-Mediated Rejection, Irrespective of Treatment-A Single Center Retrospective Study. J Clin Med 2021; 11:jcm11010199. [PMID: 35011939 PMCID: PMC8745558 DOI: 10.3390/jcm11010199] [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: 12/06/2021] [Revised: 12/25/2021] [Accepted: 12/27/2021] [Indexed: 01/16/2023] Open
Abstract
The Banff 2017 report permits the diagnosis of pure chronic antibody-mediated rejection (cAMR) in absence of microcirculation inflammation. We retrospectively investigated renal allograft function and long-term outcomes of 67 patients with cAMR, and compared patients who received antihumoral therapy (cAMR-AHT, n = 21) with patients without treatment (cAMRwo, n = 46). At baseline, the cAMR-AHT group had more concomitant T-cell-mediated rejection (9/46 (19.2%) vs. 10/21 (47.6%); p = 0.04), a higher g-lesion score (0.4 ± 0.5 versus 0.1 ± 0.3; p = 0.01) and a higher median eGFR decline in the six months prior to biopsy (6.6 vs. 3.0 mL/min; p = 0.04). The median eGFR decline six months after biopsy was comparable (2.6 vs. 4.9 mL/min, p = 0.61) between both groups, and three-year graft survival after biopsy was statistically lower in the cAMR-AHT group (35.0% vs. 61.0%, p = 0.03). Patients who received AHT had more infections (0.38 vs. 0.20 infections/patient; p = 0.04). Currently, antihumoral therapy is more often administered to patients with cAMR and rapidly deteriorating renal function or concomitant TCMR. However, long-term graft outcomes remain poor, despite treatment.
Collapse
Affiliation(s)
- Kaiyin Wu
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany; (D.S.); (C.L.d.M.); (B.O.); (Q.Z.); (F.H.); (M.C.); (F.B.); (S.R.); (W.D.); (M.G.N.); (E.S.); (K.B.)
- Correspondence: ; Tel.: +49-30-450-514002; Fax: +49-30-450-514902
| | - Danilo Schmidt
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany; (D.S.); (C.L.d.M.); (B.O.); (Q.Z.); (F.H.); (M.C.); (F.B.); (S.R.); (W.D.); (M.G.N.); (E.S.); (K.B.)
| | - Covadonga López del Moral
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany; (D.S.); (C.L.d.M.); (B.O.); (Q.Z.); (F.H.); (M.C.); (F.B.); (S.R.); (W.D.); (M.G.N.); (E.S.); (K.B.)
| | - Bilgin Osmanodja
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany; (D.S.); (C.L.d.M.); (B.O.); (Q.Z.); (F.H.); (M.C.); (F.B.); (S.R.); (W.D.); (M.G.N.); (E.S.); (K.B.)
| | - Nils Lachmann
- Institute of Transfusion Medicine, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany;
| | - Qiang Zhang
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany; (D.S.); (C.L.d.M.); (B.O.); (Q.Z.); (F.H.); (M.C.); (F.B.); (S.R.); (W.D.); (M.G.N.); (E.S.); (K.B.)
| | - Fabian Halleck
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany; (D.S.); (C.L.d.M.); (B.O.); (Q.Z.); (F.H.); (M.C.); (F.B.); (S.R.); (W.D.); (M.G.N.); (E.S.); (K.B.)
| | - Mira Choi
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany; (D.S.); (C.L.d.M.); (B.O.); (Q.Z.); (F.H.); (M.C.); (F.B.); (S.R.); (W.D.); (M.G.N.); (E.S.); (K.B.)
| | - Friederike Bachmann
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany; (D.S.); (C.L.d.M.); (B.O.); (Q.Z.); (F.H.); (M.C.); (F.B.); (S.R.); (W.D.); (M.G.N.); (E.S.); (K.B.)
| | - Simon Ronicke
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany; (D.S.); (C.L.d.M.); (B.O.); (Q.Z.); (F.H.); (M.C.); (F.B.); (S.R.); (W.D.); (M.G.N.); (E.S.); (K.B.)
| | - Wiebke Duettmann
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany; (D.S.); (C.L.d.M.); (B.O.); (Q.Z.); (F.H.); (M.C.); (F.B.); (S.R.); (W.D.); (M.G.N.); (E.S.); (K.B.)
- Berlin Institute of Health, Anna-Louisa-Karsch-Str. 2, 10178 Berlin, Germany
| | - Marcel G. Naik
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany; (D.S.); (C.L.d.M.); (B.O.); (Q.Z.); (F.H.); (M.C.); (F.B.); (S.R.); (W.D.); (M.G.N.); (E.S.); (K.B.)
- Berlin Institute of Health, Anna-Louisa-Karsch-Str. 2, 10178 Berlin, Germany
| | - Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany; (D.S.); (C.L.d.M.); (B.O.); (Q.Z.); (F.H.); (M.C.); (F.B.); (S.R.); (W.D.); (M.G.N.); (E.S.); (K.B.)
- Berlin Institute of Health, Anna-Louisa-Karsch-Str. 2, 10178 Berlin, Germany
| | - Birgit Rudolph
- Institute of Pathology, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany;
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany; (D.S.); (C.L.d.M.); (B.O.); (Q.Z.); (F.H.); (M.C.); (F.B.); (S.R.); (W.D.); (M.G.N.); (E.S.); (K.B.)
| |
Collapse
|