1
|
Chandran S, Vincenti F. Antiplasma cell antibodies: A new era of human leukocyte antigen antibody control in solid organ transplantation. Am J Transplant 2025; 25:19-26. [PMID: 39384021 DOI: 10.1016/j.ajt.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/21/2024] [Accepted: 10/03/2024] [Indexed: 10/11/2024]
Abstract
New therapies directed against plasma cells such as anti-CD38 antibodies and the bispecific anti-B cell maturation antigen antibodies, represent not only an important advance in the treatment of multiple myeloma but have the potential to change the treatment landscape of other antibody-mediated diseases. In solid organ transplantation, the therapeutic armamentarium targeting humoral alloimmune responses in desensitization of highly sensitized transplant candidates and posttransplant antibody-mediated rejection has lagged behind advances in preventing and treating T cell-mediated rejection. Intravenous immunoglobulin and plasmapheresis are used extensively but have limited efficacy. Currently available anti-CD20 antibodies are only partially effective in achieving B cell depletion and leaving mature plasma cells untouched. Although interleukin 6 plays an important role in the humoral alloimmune response and injury, the benefits of interleukin 6 inhibition have failed to be demonstrated in clinical trials. Even proteasome inhibitors developed specifically to target plasma cells have not fulfilled their promise, due to limited efficacy as single agents. This review focuses on the recent experience with, and potential applicability of, anti-CD38 antibodies in the field of organ transplantation and experimental data supporting their use and development for human leukocyte antigen desensitization and antibody-mediated rejection.
Collapse
Affiliation(s)
- Sindhu Chandran
- Cedars Sinai Comprehensive Transplant Center, Division of Nephrology, Department of Medicine, Los Angeles, CA, USA
| | - Flavio Vincenti
- Kidney Transplant Service, Departments of Medicine and Surgery, University of California-San Francisco, San Francisco, CA, USA.
| |
Collapse
|
2
|
Graham C, Litzow M. The use of haploidentical stem cell transplant as an alternative donor source in patients with decreased access to matched unrelated donors. Hematology 2024; 29:2338300. [PMID: 38753458 DOI: 10.1080/16078454.2024.2338300] [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: 12/18/2023] [Accepted: 03/28/2024] [Indexed: 05/18/2024] Open
Abstract
INTRODUCTION The likelihood of finding HLA-matched unrelated donors for rare HLA types and non-white European ancestry continues to be a challenge with less than a 70% chance of finding a full match. Mismatched transplants continue to have high rates of transplant-related mortality. With the near-universal ability to find a haploidentical donor in families, haploidentical transplants have become of more critical importance in ethnic minority groups and patients with rare HLA types. METHODS Data was collected through clinical trials, review articles, and case reports published in the National Library of Medicine. RESULTS The use of improved lymphodepleting conditioning regimens, graft versus host disease (GVHD) prophylaxis using regimens such as post-transplant cyclophosphamide, mycophenolate, and tacrolimus have improved engraftment to nearly 100 percent and reduced transplant-related mortality to less than 20 percent. Attention to donor-specific antibodies (DSAs) with interventions using bortezomib, rituximab, and plasmapheresis has decreased graft failure rates. CONCLUSION With improved prevention of GVHD with interventions such as post-transplant cyclophosphamide and management of DSAs, haploidentical transplants continue to improve transplant-related mortality (TRM) compared to patients who received matched-related donor transplants. While TRM continues to improve, ongoing research with haploidentical transplants will focus on improving graft and donor immunosuppression and identifying the best regimens to improve TRM without compromising relapse-free survival.
Collapse
Affiliation(s)
| | - Mark Litzow
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
3
|
Fattahi AS, Jafari M, Farahavar G, Abolmaali SS, Tamaddon AM. Expanding horizons in cancer therapy by immunoconjugates targeting tumor microenvironments. Crit Rev Oncol Hematol 2024; 201:104437. [PMID: 38977144 DOI: 10.1016/j.critrevonc.2024.104437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024] Open
Abstract
Immunoconjugates are promising molecules combining antibodies with different agents, such as toxins, drugs, radionuclides, or cytokines that primarily aim to target tumor cells. However, tumor microenvironment (TME), which comprises a complex network of various cells and molecular cues guiding tumor growth and progression, remains a major challenge for effective cancer therapy. Our review underscores the pivotal role of TME in cancer therapy with immunoconjugates, examining the intricate interactions with TME and recent advancements in TME-targeted immunoconjugates. We explore strategies for targeting TME components, utilizing diverse antibodies such as neutralizing, immunomodulatory, immune checkpoint inhibitors, immunostimulatory, and bispecific antibodies. Additionally, we discuss different immunoconjugates, elucidating their mechanisms of action, advantages, limitations, and applications in cancer immunotherapy. Furthermore, we highlight emerging technologies enhancing the safety and efficacy of immunoconjugates, such as antibody engineering, combination therapies, and nanotechnology. Finally, we summarize current advancements, perspectives, and future developments of TME-targeted immunoconjugates.
Collapse
Affiliation(s)
- Amir Saamaan Fattahi
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mahboobeh Jafari
- Center for Nanotechnology in Drug Delivery School of Pharmacy, Shiraz University of Medical Sciences, Iran.
| | - Ghazal Farahavar
- Center for Nanotechnology in Drug Delivery School of Pharmacy, Shiraz University of Medical Sciences, Iran.
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Center for Nanotechnology in Drug Delivery School of Pharmacy, Shiraz University of Medical Sciences, Iran.
| | - Ali Mohammad Tamaddon
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Center for Nanotechnology in Drug Delivery School of Pharmacy, Shiraz University of Medical Sciences, Iran.
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Viklicky O, Hruba P, Novotny M, Kment M, Roder M, Halloran PF, Böhmig GA. Targeting CD38 in Subclinical Antibody-mediated Rejection in HLA-incompatible Kidney Transplantation: A Case Report. Transplant Direct 2024; 10:e1685. [PMID: 39035116 PMCID: PMC11259386 DOI: 10.1097/txd.0000000000001685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/15/2024] [Accepted: 06/02/2024] [Indexed: 07/23/2024] Open
Affiliation(s)
- Ondrej Viklicky
- Department of Nephrology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Transplant Laboratory, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Petra Hruba
- Transplant Laboratory, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Marek Novotny
- Department of Nephrology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Martin Kment
- Department of Pathology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Matej Roder
- Department of Immunogenetics, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | | | - Georg A. Böhmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna, Vienna, Austria
| |
Collapse
|
7
|
Mohidin B, Needleman A, Fernando R, Lowe DM, Wechalekar A, Sheaff M, Salama A, Jones G. Renal Transplant Outcomes in Plasma Cell Dyscrasias and AL Amyloidosis after Treatment with Daratumumab. J Clin Med 2024; 13:4109. [PMID: 39064149 PMCID: PMC11278235 DOI: 10.3390/jcm13144109] [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: 06/10/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Background: The morbidity and mortality from AL amyloidosis has significantly improved with the development of novel treatments. Daratumumab is a highly effective treatment for AL amyloidosis, but end-stage kidney disease is a common complication of this condition. Kidney transplantation is the ideal form of renal replacement therapy but has historically been contraindicated in this group of patients. Methods: Given the improved survival and better treatments of both conditions, we argue that it is time to reconsider transplanting these patients. Results: We report our experience of transplanting four patients with AL amyloidosis who had achieved stable remission through treatment with daratumumab. Conclusions: We highlight the key challenges involved and discuss important clinical issues for patients receiving daratumumab, particularly the difficulties with interpreting the crossmatch in light of daratumumab and immunoglobulin therapy interference. We also discuss the complexities involved in balancing the risks of infection, relapse, rejection, and immunosuppression in such patients.
Collapse
Affiliation(s)
- Barian Mohidin
- UCL Department of Renal Medicine, Royal Free Hospital, London NW3 2QG, UK
| | - Amy Needleman
- UCL Department of Renal Medicine, Royal Free Hospital, London NW3 2QG, UK
| | | | - David M. Lowe
- UCL Institute of Immunity & Transplantation, Royal Free Hospital, London NW3 2QG, UK
| | | | - Michael Sheaff
- Department of Cellular Pathology, Royal London Hospital, London NW3 2QG, UK
| | - Alan Salama
- UCL Department of Renal Medicine, Royal Free Hospital, London NW3 2QG, UK
| | - Gareth Jones
- UCL Department of Renal Medicine, Royal Free Hospital, London NW3 2QG, UK
| |
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
|
Rroji M, Figurek A, Spasovski G. Advancing kidney transplant outcomes: the role of urinary proteomics in graft function monitoring and rejection detection. Expert Rev Proteomics 2024; 21:297-316. [PMID: 39133121 DOI: 10.1080/14789450.2024.2389829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/15/2024] [Accepted: 07/30/2024] [Indexed: 08/13/2024]
Abstract
INTRODUCTION Kidney transplantation significantly improves the lives of those with end-stage kidney disease, offering best alternative to dialysis. However, transplant success is threatened by the acute and chronic rejection mechanisms due to complex immune responses against the new organ. AREAS COVERED The ongoing research into biomarkers holds promise for revolutionizing the early detection and monitoring of the graft health. Liquid biopsy techniques offer a new avenue, with several diagnostic, predictive, and prognostic biomarkers showing promise in detecting and monitoring kidney diseases and an early and chronic allograft rejection. EXPERT OPINION Evaluating the protein composition related to kidney transplant results could lead to identifying biomarkers that provide insights into the graft functionality. Non-invasive proteomic biomarkers can drastically enhance clinical outcomes and change the way how kidney transplants are evaluated for patients and physicians if they succeed in this transition. Hence, the advancement in proteomic technologies, leads toward a significant improvement in understanding of the protein markers and molecular mechanisms linked to the outcomes of kidney transplants. However, the road from discovery to the use of such proteins in clinical practice is long, with a need for continuous validation and beyond the singular research team with comprehensive infrastructure and across research groups collaboration.
Collapse
Affiliation(s)
- Merita Rroji
- Faculty of Medicine, University Department of Nephrology, University of Medicine Tirana, Tirana, Albania
| | - Andreja Figurek
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Goce Spasovski
- Medical Faculty, University Department of Nephrology, University of Skopje, Skopje, Macedonia
| |
Collapse
|
10
|
Diebold M, Farkash EA, Barnes J, Regele H, Kozakowski N, Schatzl M, Mayer KA, Haindl S, Vietzen H, Hidalgo LG, Halloran PF, Eskandary F, Böhmig GA. Natural Killer Cell Presence in Antibody-Mediated Rejection. Transpl Int 2024; 37:13209. [PMID: 38979120 PMCID: PMC11228143 DOI: 10.3389/ti.2024.13209] [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: 06/12/2024] [Indexed: 07/10/2024]
Abstract
Transcript analyses highlight an important contribution of natural killer (NK) cells to microvascular inflammation (MVI) in antibody-mediated rejection (ABMR), but only few immunohistologic studies have quantified their spatial distribution within graft tissue. This study included 86 kidney transplant recipients who underwent allograft biopsies for a positive donor-specific antibody (DSA) result. NK cells were visualized and quantified within glomeruli and peritubular capillaries (PTC), using immunohistochemistry for CD34 alongside CD16/T-bet double-staining. Staining results were analyzed in relation to histomorphology, microarray analysis utilizing the Molecular Microscope Diagnostic System, functional NK cell genetics, and clinical outcomes. The number of NK cells in glomeruli per mm2 glomerular area (NKglom) and PTC per mm2 cortical area (NKPTC) was substantially higher in biopsies with ABMR compared to those without rejection, and correlated with MVI scores (NKglom Spearman's correlation coefficient [SCC] = 0.55, p < 0.001, NKPTC 0.69, p < 0.001). In parallel, NK cell counts correlated with molecular classifiers reflecting ABMR activity (ABMRprob: NKglom 0.59, NKPTC 0.75) and showed a trend towards higher levels in association with high functional FCGR3A and KLRC2 gene variants. Only NKPTC showed a marginally significant association with allograft function and survival. Our immunohistochemical results support the abundance of NK cells in DSA-positive ABMR.
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, Basel, Switzerland
| | - Evan A Farkash
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Jenna Barnes
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Heinz Regele
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | | | - Martina Schatzl
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Katharina A Mayer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Susanne Haindl
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Hannes Vietzen
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Luis G Hidalgo
- Department of Surgery, University of Wisconsin, Madison, WI, United States
| | - Philip F Halloran
- Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Farsad Eskandary
- 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
|
11
|
Schrezenmeier E, Choi M, Globke B, Dörner T, Leimbach A, Osmanodja B, Schramm A, Amann K, Eckardt KU, Budde K, Öllinger R, Lachmann N, Halleck F. Successful Desensitization with Imlifidase and Daratumumab in a Highly Immunized, Crossmatch Positive, Blood Group-Incompatible Living-Donor Re-Transplant Recipient with Systemic Lupus Erythematosus and Antiphospholipid Syndrome. Transfus Med Hemother 2024; 51:158-163. [PMID: 38867806 PMCID: PMC11166406 DOI: 10.1159/000538513] [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: 02/06/2024] [Accepted: 03/19/2024] [Indexed: 06/14/2024] Open
Abstract
Introduction The transplantation of highly sensitized patients remains a major obstacle. Immunized patients wait longer for a transplant if not prioritized, and if transplanted, their transplant outcome is worse. Case Presentation We report a successful AB0- and HLA-incompatible living donor kidney transplantation in a 35-year-old female patient with systemic lupus erythematosus (SLE) and antiphospholipid syndrome. The patient had a positive T- and B-cell complement-dependent cytotoxicity (CDC) crossmatch and previous graft loss due to renal vein thrombosis. We treated the patient with intravenous immunoglobulins, rituximab, horse anti-thymocyte globulin, daratumumab, and imlifidase, besides standard immunosuppression. All IgG antibodies were sensitive to imlifidase treatment. Besides donor-specific HLA antibodies, anti-dsDNA antibodies and antiphospholipid antibodies were cleaved. The patient initially had delayed graft function. Two kidney biopsies (day 7 and day 14) revealed acute tubular necrosis without signs of HLA antibody-mediated rejection. On posttransplant day 30, hemodialysis was stopped, and creatinine levels declined over the next weeks to a baseline creatinine of about 1.7 mg/dL after 12 months. Conclusion In this case, a novel multimodal treatment strategy including daratumumab and imlifidase enabled successful kidney transplantation for a highly immunized patient with antiphospholipid antibodies.
Collapse
Affiliation(s)
- Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health Charité Universitätsmedizin Berlin, Berlin Institute of Health (BIH) Academy, Berlin, Germany
| | - Mira Choi
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Brigitta Globke
- Berlin Institute of Health Charité Universitätsmedizin Berlin, Berlin Institute of Health (BIH) Academy, Berlin, Germany
- Department of Surgery Charité Campus Mitte, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Thomas Dörner
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Deutsches Rheuma-Forschungszentrum, A Leibniz Institute, Berlin, Germany
| | - Alexandra Leimbach
- Department of Surgery Charité Campus Mitte, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Bilgin Osmanodja
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Alexander Schramm
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Kerstin Amann
- Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Robert Öllinger
- Department of Surgery Charité Campus Mitte, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Nils Lachmann
- Institute for Transfusion Medicine, HLA-Laboratory, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Fabian Halleck
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
12
|
Chen Z, Xu Q, Shou Z. Application of CD38 monoclonal antibody in kidney disease. Front Immunol 2024; 15:1382977. [PMID: 38799465 PMCID: PMC11116655 DOI: 10.3389/fimmu.2024.1382977] [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: 02/06/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024] Open
Abstract
CD38 antigen is a glycoprotein that found on the surface of several immune cells, and this property makes its monoclonal antibodies have the effect of targeted elimination of immune cells. Therefore, the CD38 monoclonal antibody (such as daratumumab, Isatuximab) becomes a new treatment option for membranous nephropathy, lupus nephritis, renal transplantation, and other refractory kidney diseases. This review summarizes the application of CD38 monoclonal antibodies in different kidney diseases and highlights future prospects.
Collapse
Affiliation(s)
- Zhiyi Chen
- College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Nephrology, Shulan (Hangzhou) Hospital, Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang, China
| | - Qianchun Xu
- Department of Nephrology, Shulan (Hangzhou) Hospital, Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang, China
- Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Zhangfei Shou
- Department of Nephrology, Shulan (Hangzhou) Hospital, Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
13
|
de Nattes T, Beadle J, Roufosse C. Biopsy-based transcriptomics in the diagnosis of kidney transplant rejection. Curr Opin Nephrol Hypertens 2024; 33:273-282. [PMID: 38411022 PMCID: PMC10990030 DOI: 10.1097/mnh.0000000000000974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
PURPOSE OF REVIEW The last year has seen considerable progress in translational research exploring the clinical utility of biopsy-based transcriptomics of kidney transplant biopsies to enhance the diagnosis of rejection. This review will summarize recent findings with a focus on different platforms, potential clinical applications, and barriers to clinical adoption. RECENT FINDINGS Recent literature has focussed on using biopsy-based transcriptomics to improve diagnosis of rejection, in particular antibody-mediated rejection. Different techniques of gene expression analysis (reverse transcriptase quantitative PCR, microarrays, probe-based techniques) have been used either on separate samples with ideally preserved RNA, or on left over tissue from routine biopsy processing. Despite remarkable consistency in overall patterns of gene expression, there is no consensus on acceptable indications, or whether biopsy-based transcriptomics adds significant value at reasonable cost to current diagnostic practice. SUMMARY Access to biopsy-based transcriptomics will widen as regulatory approvals for platforms and gene expression models develop. Clinicians need more evidence and guidance to inform decisions on how to use precious biopsy samples for biopsy-based transcriptomics, and how to integrate results with standard histology-based diagnosis.
Collapse
Affiliation(s)
- Tristan de Nattes
- Univ Rouen Normandie, INSERM U1234, CHU Rouen, Department of Nephrology, Rouen, France
| | - Jack Beadle
- Centre for Inflammatory Diseases, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Candice Roufosse
- Centre for Inflammatory Diseases, Department of Immunology and Inflammation, Imperial College London, London, UK
| |
Collapse
|
14
|
Provenzano M, Hu L, Tringali E, Senatore M, Talarico R, Di Dio M, Ruotolo C, La Manna G, Garofalo C, Zaza G. Improving Kidney Disease Care: One Giant Leap for Nephrology. Biomedicines 2024; 12:828. [PMID: 38672183 PMCID: PMC11048002 DOI: 10.3390/biomedicines12040828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Nephrology is an ever-evolving field of medicine. The importance of such a discipline is related to the high clinical impact of kidney disease. In fact, abnormalities of kidney function and/or structure are common in the general population, reaching an overall prevalence of about 10%. More importantly, the onset of kidney damage is related to a strikingly high risk of cardiovascular events, mortality, and progression to kidney failure which, in turn, compromises quality and duration of life. Attempts to comprehend the pathogenesis and molecular mechanisms involved in kidney disease occurrence have prompted the development and implementation of novel drugs in clinical practice with the aim of treating the 'specific cause' of kidney disease (including chronic kidney disease, glomerular disease, and genetic kidney disorders) and the main immunological complications following kidney transplantation. Herein, we provide an overview of the principal emerging drug classes with proved efficacy in the context of the aforementioned clinical conditions. This can represent a simplified guide for clinical nephrologists to remind them of the vast and heterogeneous armamentarium of drugs that should be used in the present and the future to improve the management of patients suffering from kidney disease.
Collapse
Affiliation(s)
- Michele Provenzano
- Department of Pharmacy Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (M.S.); (R.T.)
| | - Lilio Hu
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (L.H.); (E.T.); (G.L.M.)
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy
| | - Edoardo Tringali
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (L.H.); (E.T.); (G.L.M.)
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy
| | - Massimo Senatore
- Department of Pharmacy Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (M.S.); (R.T.)
| | - Roberta Talarico
- Department of Pharmacy Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (M.S.); (R.T.)
| | - Michele Di Dio
- Division of Urology, Department of Surgery, SS Annunziata Hospital, 87100 Cosenza, Italy;
| | - Chiara Ruotolo
- Unit of Nephrology, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (C.R.); (C.G.)
| | - Gaetano La Manna
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (L.H.); (E.T.); (G.L.M.)
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy
| | - Carlo Garofalo
- Unit of Nephrology, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (C.R.); (C.G.)
| | - Gianluigi Zaza
- Department of Pharmacy Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (M.S.); (R.T.)
| |
Collapse
|
15
|
Heeger PS, Haro MC, Jordan S. Translating B cell immunology to the treatment of antibody-mediated allograft rejection. Nat Rev Nephrol 2024; 20:218-232. [PMID: 38168662 DOI: 10.1038/s41581-023-00791-0] [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] [Accepted: 11/10/2023] [Indexed: 01/05/2024]
Abstract
Antibody-mediated rejection (AMR), including chronic AMR (cAMR), causes ~50% of kidney allograft losses each year. Despite attempts to develop well-tolerated and effective therapeutics for the management of AMR, to date, none has obtained FDA approval, thereby highlighting an urgent unmet medical need. Discoveries over the past decade from basic, translational and clinical studies of transplant recipients have provided a foundation for developing novel therapeutic approaches to preventing and treating AMR and cAMR. These interventions are aimed at reducing donor-specific antibody levels, decreasing graft injury and fibrosis, and preserving kidney function. Innovative approaches emerging from basic science findings include targeting interactions between alloreactive T cells and B cells, and depleting alloreactive memory B cells, as well as donor-specific antibody-producing plasmablasts and plasma cells. Therapies aimed at reducing the cytotoxic antibody effector functions mediated by natural killer cells and the complement system, and their associated pro-inflammatory cytokines, are also undergoing evaluation. The complexity of the pathogenesis of AMR and cAMR suggest that multiple approaches will probably be required to treat these disease processes effectively. Definitive answers await results from large, double-blind, multicentre, randomized controlled clinical trials.
Collapse
Affiliation(s)
- Peter S Heeger
- Comprehensive Transplant Center, Department of Medicine, Division of Nephrology Cedars-Sinai Medical Center Los Angeles, Los Angeles, CA, USA
| | - Maria Carrera Haro
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, Mount Sinai, NY, USA
| | - Stanley Jordan
- Comprehensive Transplant Center, Department of Medicine, Division of Nephrology Cedars-Sinai Medical Center Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
16
|
Lemal R, Blandin L, Uro-Coste C, Philipponnet C, Geoffroy E, Heng AE, Garrouste C, Rouzaire P. Daratumumab treatment in six highly sensitised solid organ transplant recipients: A case series and literature review. HLA 2024; 103:e15458. [PMID: 38597238 DOI: 10.1111/tan.15458] [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: 12/19/2023] [Revised: 03/04/2024] [Accepted: 03/15/2024] [Indexed: 04/11/2024]
Abstract
We report data on six kidney or heart recipients who were administered daratumumab to treat or prevent antibody-mediated rejection (ABMR). To date, data are scarce concerning the use of daratumumab in solid organ transplantation and most reports show a decrease in donor-specific antigen (DSA) levels and an improvement in ABMR using a multiple myeloma daratumumab administration scheme, that is, with sequential systematic administration. Here, we report on the efficacy of daratumumab 1/ in reducing the histological signs of ABMR, 2/ in reducing the ability of DSA to bind to donor cells in vitro through negativation of flow cytometry crossmatching, 3/ in preferentially being directed towards antibodies sharing epitopes, suggesting that daratumumab may specifically target activated plasma cells, 4/ and when administered as a single dose. This last point suggests, for the first time, that, as for rituximab in auto-immune diseases, the scheme for daratumumab administration could be different for targeting DSA-producing plasma cells than for tumour cells.
Collapse
Affiliation(s)
- Richard Lemal
- Service d'Histocompatibilité et Immunogénétique, Centre Hospitalo-Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
- EA 7453 CHELTER, Université Clermont Auvergne UCA, Clermont-Ferrand, France
| | - Lucie Blandin
- Service d'Histocompatibilité et Immunogénétique, Centre Hospitalo-Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Charlotte Uro-Coste
- Service de Néphrologie, Centre Hospitalo-Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Carole Philipponnet
- Service de Néphrologie, Centre Hospitalo-Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Etienne Geoffroy
- Service de Cardiologie, Centre Hospitalo-Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Anne-Elisabeth Heng
- Service de Néphrologie, Centre Hospitalo-Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Cyril Garrouste
- EA 7453 CHELTER, Université Clermont Auvergne UCA, Clermont-Ferrand, France
- Service de Néphrologie, Centre Hospitalo-Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Paul Rouzaire
- Service d'Histocompatibilité et Immunogénétique, Centre Hospitalo-Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
- EA 7453 CHELTER, Université Clermont Auvergne UCA, Clermont-Ferrand, France
| |
Collapse
|
17
|
Angeletti A, Bin S, Magnasco A, Bruschi M, Cravedi P, Ghiggeri GM. Efficacy of combined rituximab and daratumumab treatment in posttransplant recurrent focal segmental glomerulosclerosis. Am J Transplant 2024; 24:688-692. [PMID: 38101474 DOI: 10.1016/j.ajt.2023.12.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/15/2023] [Accepted: 12/01/2023] [Indexed: 12/17/2023]
Abstract
Focal segmental glomerulosclerosis (FSGS) is one of the leading causes of kidney failure and it is characterized by a high rate of recurrence after kidney transplant. Moreover, FSGS recurrence is worsened by an increased risk of graft failure. Common therapies for FSGS recurrence mostly consist of plasma exchange treatments, also for prolonged time, and rituximab, with variable efficacy. We report 5 cases of early FSGS recurrence after kidney transplant, resistant to plasma exchange and rituximab treatment that subsequently resolved after combined therapy with rituximab and daratumumab. All cases were negative for genetic FSGS. The combined treatment induced a complete response in all the cases and was well tolerated. We also performed a comprehensive flow cytometry analysis in 2 subjects that may suggest a mechanistic link between plasma cells and disease activity. In conclusion, given the lack of viable treatments for recurrent FSGS, our reports support the rationale for a pilot trial testing the safety/efficacy profile of combined rituximab and daratumumab in posttransplant FSGS recurrence.
Collapse
Affiliation(s)
- Andrea Angeletti
- Nephrology, Dialysis and Transplantation Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy.
| | - Sofia Bin
- Translational Transplant Research Center and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Alberto Magnasco
- Nephrology, Dialysis and Transplantation Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Maurizio Bruschi
- Nephrology, Dialysis and Transplantation Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Paolo Cravedi
- Translational Transplant Research Center and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Gian Marco Ghiggeri
- Nephrology, Dialysis and Transplantation Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
18
|
Reindl-Schwaighofer R, Heinzel A, Oberbauer R. Continuous surveillance for kidney transplant rejection: ready for clinical trials? Clin Kidney J 2024; 17:sfad242. [PMID: 38495221 PMCID: PMC10941637 DOI: 10.1093/ckj/sfad242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Indexed: 03/19/2024] Open
Affiliation(s)
| | - Andreas Heinzel
- Department of Nephrology, Medical University of Vienna, Vienna, Austria
| | - Rainer Oberbauer
- Department of Nephrology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
19
|
Leung N, Heybeli C. Kidney Transplantation in Multiple Myeloma and Monoclonal Gammopathy of Renal Significance. Semin Nephrol 2024; 44:151497. [PMID: 38485643 DOI: 10.1016/j.semnephrol.2024.151497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
Recent advances in the treatment of plasma cell disorders (PCDs) have provided a wealth of therapy alternatives and improved overall survival tremendously. Various types of PCDs are associated with kidney injury and end-stage kidney disease in a considerable number of patients. Kidney transplantation (KTx) is the best option for renal replacement therapy in select patients in terms of both quality of life parameters and overall survival. Even with modern therapies, all PCDs carry the risk of hematologic progression, whereas histologic recurrence and graft loss are other prevailing concerns in these patients. The risk of mortality is also higher in some of these disorders compared with KTx recipients who suffer from other causes of kidney disease. Unlike solid cancers, there is no well-defined "waiting time" after hematologic remission before proceeding to KTx. Thus, clinicians are usually reluctant to recommend KTx to patients who develop end-stage kidney disease due to PCDs. This review aims to provide the current evidence on KTx outcomes in patients with monoclonal gammopathy of renal significance and multiple myeloma. Although immunoglobulin light chain amyloidosis is a monoclonal gammopathy of renal significance subtype, KTx outcomes in this group are mentioned in another chapter of this issue.
Collapse
Affiliation(s)
- Nelson Leung
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN; Division of Hematology, Mayo Clinic, Rochester, MN.
| | - Cihan Heybeli
- Division of Nephrology, Dokuz Eylül University Hospital, Balcova, Turkey
| |
Collapse
|
20
|
de Nattes T, Kaveri R, Farce F, François A, Guerrot D, Hanoy M, Laurent C, Candon S, Bertrand D. Daratumumab for antibody-mediated rejection: Is it time to target the real culprit? Am J Transplant 2023; 23:1990-1994. [PMID: 37414251 DOI: 10.1016/j.ajt.2023.06.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/08/2023]
Abstract
We report the case of a sensitized woman who underwent successful transplantation after a desensitization protocol, with an optically normal 8-day biopsy. At 3 months, she developed active antibody-mediated rejection (AMR) due to preformed donor-specific antibodies. It was decided to treat the patient with daratumumab, an anti-CD38 monoclonal antibody. The mean fluorescence intensity of donor-specific antibodies decreased, pathologic signs of AMR regressed, and kidney function returned to normal. A molecular assessment of biopsies was retrospectively performed. By doing so, regression of the molecular signature of AMR was evidenced between the second and third biopsies. Interestingly, the first biopsy revealed a gene expression profile of AMR, which helped retrospectively classify this biopsy as AMR, illustrating the relevance of molecular phenotyping of biopsy in high-risk situations such as desensitization.
Collapse
Affiliation(s)
- Tristan de Nattes
- Univ Rouen Normandie, INSERM U1234, CHU Rouen, Service de Néphrologie, Rouen, France.
| | - Rangolie Kaveri
- EFS Hauts de France-Normandie, Service d'Histocompatibilité, Rouen, France
| | - Fabienne Farce
- EFS Hauts de France-Normandie, Service d'Histocompatibilité, Rouen, France
| | | | - Dominique Guerrot
- Univ Rouen Normandie, INSERM U1096, CHU Rouen, CIC-CRB 1404, Service de Néphrologie, Rouen, France
| | | | | | - Sophie Candon
- Univ Rouen Normandie, INSERM U1234, CHU Rouen, Service d'Immunologie et de Biothérapies, Rouen, France
| | | |
Collapse
|
21
|
Süsal CC, Kraft L, Ender A, Süsal C, Schwenger A, Amann K, Böhmig GA, Schwenger V. Blood group-specific apheresis in combination with daratumumab as a rescue therapy of acute antibody-mediated rejection in a case of ABO- and human leukocyte antigen-incompatible kidney transplantation. SAGE Open Med Case Rep 2023; 11:2050313X231211050. [PMID: 38022864 PMCID: PMC10631334 DOI: 10.1177/2050313x231211050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
We report a case of antibody-mediated rejection treated with the human CD38 monoclonal antibody daratumumab in a 58-year-old female patient with end-stage kidney disease due to autosomal dominant polycystic kidney disease who received an ABO- and human leukocyte antigen antibody-incompatible living donor kidney transplant. The patient experienced an episode of severe antibody-mediated rejection within the first week of transplantation. Blood-group-antibody selective immunoadsorption in combination with administration of four doses of daratumumab (each 1800 mg s.c.) led to a persistent decrease of ABO- and more interestingly donor-specific human leukocyte antigen antibody reactivity and resulted in clinical and histopathological remission with full recovery of graft function, which has remained stable until post-transplant day 212. This case illustrates the potential of targeting CD38 in antibody-mediated rejection.
Collapse
Affiliation(s)
- Can C Süsal
- Department of Nephrology, Klinikum Stuttgart—Katharinenhospital, Stuttgart, Germany
| | - Leonie Kraft
- Department of Nephrology, Klinikum Stuttgart—Katharinenhospital, Stuttgart, Germany
| | - Andrea Ender
- Central Institute for Transfusion Medicine and Blood Donation, Katharinenhospital Stuttgart, Stuttgart, Germany
| | - Caner Süsal
- Transplant Immunology Research Center of Excellence, Koç University Hospital, Istanbul, Turkey
| | - Amelie Schwenger
- Experimental Immunology, Department for Children and Adolescents Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Kerstin Amann
- Department of Nephropathology, Department of Pathology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Georg A Böhmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Vedat Schwenger
- Department of Nephrology, Klinikum Stuttgart—Katharinenhospital, Stuttgart, Germany
| |
Collapse
|
22
|
Brandon W, Dunn C, Bollineni S, Joerns J, Lawrence A, Mohanka M, Timofte I, Torres F, Kaza V. Management of donor-specific antibodies in lung transplantation. FRONTIERS IN TRANSPLANTATION 2023; 2:1248284. [PMID: 38993917 PMCID: PMC11235237 DOI: 10.3389/frtra.2023.1248284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/23/2023] [Indexed: 07/13/2024]
Abstract
The formation of antibodies against donor human leukocyte antigens poses a challenging problem both for donor selection as well as postoperative graft function in lung transplantation. These donor-specific antibodies limit the pool of potential donor organs and are associated with episodes of antibody-mediated rejection, chronic lung allograft dysfunction, and increased mortality. Optimal management strategies for clearance of DSAs are poorly defined and vary greatly by institution; most of the data supporting any particular strategy is limited to small-scale retrospective cohort studies. A typical approach to antibody depletion may involve the use of high-dose steroids, plasma exchange, intravenous immunoglobulin, and possibly other immunomodulators or small-molecule therapies. This review seeks to define the current understanding of the significance of DSAs in lung transplantation and outline the literature supporting strategies for their management.
Collapse
Affiliation(s)
- William Brandon
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Colin Dunn
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Srinivas Bollineni
- Division of Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - John Joerns
- Division of Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Adrian Lawrence
- Division of Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Manish Mohanka
- Division of Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Irina Timofte
- Division of Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Fernando Torres
- Division of Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Vaidehi Kaza
- Division of Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
23
|
Zhang J, Yu X, Xie Z, Wang R, Li H, Tang Z, Na N. A bibliometric and knowledge-map analysis of antibody-mediated rejection in kidney transplantation. Ren Fail 2023; 45:2257804. [PMID: 37724568 PMCID: PMC10512841 DOI: 10.1080/0886022x.2023.2257804] [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: 04/12/2023] [Accepted: 09/06/2023] [Indexed: 09/21/2023] Open
Abstract
OBJECTIVES Antibody-mediated rejection (AMR) is a large obstacle to the long-term survival of allograft kidneys. It is urgent to find novel strategies for its prevention and treatment. Bibliometric analysis is helpful in understanding the directions of one field. Hence, this study aims to analyze the state and emerging trends of AMR in kidney transplantation. METHODS Literature on AMR in kidney transplantation from 1999 to 2022 was collected from the Web of Science Core Collection. HistCite (version 12.03.17), CiteSpace (version 6.2.R2), Bibliometrix 4.1.0 Package from R language, and Gephi (https://gephi.org) were applied to the bibliometric analysis of the annual publications, leading countries/regions, core journals, references, keywords, and trend topics. RESULTS A total of 2522 articles related to AMR in kidney transplantation were included in the analysis and the annual publications increased year by year. There were 10874 authors from 118 institutions located in 70 countries/regions contributing to AMR studies, and the United States took the leading position in both articles and citation scores. Halloran PF from Canada made the most contribution to AMR in kidney transplantation. The top 3 productive journals, American Journal of Transplantation, Transplantation, and Transplantation Proceedings, were associated with transplantation. Moreover, the recent trend topics mainly focused on transplant outcomes, survival, and clinical research. CONCLUSIONS North American and European countries/regions played central roles in AMR of kidney transplantation. Importantly, the prognosis of AMR is the hotspot in the future. Noninvasive strategies like plasma and urine dd-cfDNA may be the most potential direction in the AMR field.
Collapse
Affiliation(s)
- Jinhua Zhang
- Department of kidney transplantation, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaowen Yu
- Department of General Surgery, Kunming Municipal Hospital of Traditional Chinese Medicine, the Third Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming, China
| | - Zhenwei Xie
- Department of kidney transplantation, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ruojiao Wang
- Department of kidney transplantation, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Heng Li
- Department of kidney transplantation, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - ZuoFu Tang
- Department of kidney transplantation, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ning Na
- Department of kidney transplantation, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
24
|
Lamarthée B, Callemeyn J, Van Herck Y, Antoranz A, Anglicheau D, Boada P, Becker JU, Debyser T, De Smet F, De Vusser K, Eloudzeri M, Franken A, Gwinner W, Koshy P, Kuypers D, Lambrechts D, Marquet P, Mathias V, Rabant M, Sarwal MM, Senev A, Sigdel TK, Sprangers B, Thaunat O, Tinel C, Van Brussel T, Van Craenenbroeck A, Van Loon E, Vaulet T, Bosisio F, Naesens M. Transcriptional and spatial profiling of the kidney allograft unravels a central role for FcyRIII+ innate immune cells in rejection. Nat Commun 2023; 14:4359. [PMID: 37468466 DOI: 10.1038/s41467-023-39859-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 06/28/2023] [Indexed: 07/21/2023] Open
Abstract
Rejection remains the main cause of premature graft loss after kidney transplantation, despite the use of potent immunosuppression. This highlights the need to better understand the composition and the cell-to-cell interactions of the alloreactive inflammatory infiltrate. Here, we performed droplet-based single-cell RNA sequencing of 35,152 transcriptomes from 16 kidney transplant biopsies with varying phenotypes and severities of rejection and without rejection, and identified cell-type specific gene expression signatures for deconvolution of bulk tissue. A specific association was identified between recipient-derived FCGR3A+ monocytes, FCGR3A+ NK cells and the severity of intragraft inflammation. Activated FCGR3A+ monocytes overexpressed CD47 and LILR genes and increased paracrine signaling pathways promoting T cell infiltration. FCGR3A+ NK cells overexpressed FCRL3, suggesting that antibody-dependent cytotoxicity is a central mechanism of NK-cell mediated graft injury. Multiplexed immunofluorescence using 38 markers on 18 independent biopsy slides confirmed this role of FcγRIII+ NK and FcγRIII+ nonclassical monocytes in antibody-mediated rejection, with specificity to the glomerular area. These results highlight the central involvement of innate immune cells in the pathogenesis of allograft rejection and identify several potential therapeutic targets that might improve allograft longevity.
Collapse
Affiliation(s)
- Baptiste Lamarthée
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, KU Leuven, Leuven, Belgium
- Université de Franche-Comté, UBFC, EFS, Inserm UMR RIGHT, Besançon, France
| | - Jasper Callemeyn
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, KU Leuven, Leuven, Belgium
- Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Yannick Van Herck
- Department of Oncology, Laboratory for Experimental Oncology, KU Leuven, Leuven, Belgium
| | - Asier Antoranz
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven, Leuven, Belgium
| | - Dany Anglicheau
- Department of Nephrology and Kidney Transplantation, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- Université Paris Cité, Inserm U1151, Necker Enfants-Malades Institute, Paris, France
| | - Patrick Boada
- Division of Multi-Organ Transplantation, Department of Surgery, UCSF, 513 Parnassus, San Francisco, CA, USA
| | - Jan Ulrich Becker
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Tim Debyser
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, KU Leuven, Leuven, Belgium
| | - Frederik De Smet
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven, Leuven, Belgium
| | - Katrien De Vusser
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, KU Leuven, Leuven, Belgium
- Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Maëva Eloudzeri
- Université Paris Cité, Inserm U1151, Necker Enfants-Malades Institute, Paris, France
| | - Amelie Franken
- VIB Center for Cancer Biology, Leuven, Belgium
- Department of Human Genetics, Laboratory of Translational Genetics, KU Leuven, Leuven, Belgium
| | - Wilfried Gwinner
- Department of Nephrology, Hannover Medical School, Hannover, Germany
| | - Priyanka Koshy
- Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Dirk Kuypers
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, KU Leuven, Leuven, Belgium
- Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Diether Lambrechts
- VIB Center for Cancer Biology, Leuven, Belgium
- Department of Human Genetics, Laboratory of Translational Genetics, KU Leuven, Leuven, Belgium
| | - Pierre Marquet
- Department of Pharmacology and Transplantation, University of Limoges, Inserm U1248, Limoges University Hospital, Limoges, France
| | - Virginie Mathias
- EFS, HLA Laboratory, Décines, France
- Université Claude Bernard Lyon I, Inserm U1111, CNRS UMR5308, CIRI, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Marion Rabant
- Université Paris Cité, Inserm U1151, Necker Enfants-Malades Institute, Paris, France
- Department of Pathology, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Minnie M Sarwal
- Division of Multi-Organ Transplantation, Department of Surgery, UCSF, 513 Parnassus, San Francisco, CA, USA
| | - Aleksandar Senev
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, KU Leuven, Leuven, Belgium
- Histocompatibility and Immunogenetics Laboratory, Red Cross-Flanders, Mechelen, Belgium
| | - Tara K Sigdel
- Division of Multi-Organ Transplantation, Department of Surgery, UCSF, 513 Parnassus, San Francisco, CA, USA
| | - Ben Sprangers
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, KU Leuven, Leuven, Belgium
- Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Olivier Thaunat
- Université Claude Bernard Lyon I, Inserm U1111, CNRS UMR5308, CIRI, Ecole Normale Supérieure de Lyon, Lyon, France
- Hospices Civils de Lyon, Edouard Herriot Hospital, Department of Transplantation, Nephrology and Clinical Immunology, Lyon, France
| | - Claire Tinel
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, KU Leuven, Leuven, Belgium
- Université de Franche-Comté, UBFC, EFS, Inserm UMR RIGHT, Besançon, France
- Department of Nephrology and Kidney Transplantation, Dijon Hospital, Dijon, France
| | - Thomas Van Brussel
- VIB Center for Cancer Biology, Leuven, Belgium
- Department of Human Genetics, Laboratory of Translational Genetics, KU Leuven, Leuven, Belgium
| | - Amaryllis Van Craenenbroeck
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, KU Leuven, Leuven, Belgium
- Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Elisabet Van Loon
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, KU Leuven, Leuven, Belgium
- Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Thibaut Vaulet
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, KU Leuven, Leuven, Belgium
| | - Francesca Bosisio
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven, Leuven, Belgium
| | - Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, KU Leuven, Leuven, Belgium.
- Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
25
|
Scalzo RE, Sanoff SL, Rege AS, Kwun J, Knechtle SJ, Barisoni L, Byrns JS. Daratumumab Use Prior to Kidney Transplant and T Cell-Mediated Rejection: A Case Report. Am J Kidney Dis 2023; 81:616-620. [PMID: 36623683 DOI: 10.1053/j.ajkd.2022.11.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 11/11/2022] [Indexed: 01/09/2023]
Abstract
There is growing interest in daratumumab in the solid organ transplant realm owing to the potential immunomodulatory effects on CD38-expressing cells, primarily plasma cells, as they have a key role in antibody production. In particular there is interest in use of daratumumab for desensitization and potential treatment for antibody-mediated rejection. However, ongoing investigation with daratumumab has shown potential immunologic concerns in vitro, with a significant increase in populations of CD4-positive cytotoxic T cells and CD8-positive helper T cells in both peripheral blood and bone marrow that could lead to acute T cell-mediated rejection in the solid organ transplant patient. To date, there are no published reports of an association with daratumumab use and T cell-mediated rejection in vivo. In this case report we present what is to our knowledge the first documented case of an early severe T cell-mediated rejection in a low-immunologic-risk living-donor kidney transplant recipient who received daratumumab for multiple myeloma maintenance prior to transplant.
Collapse
Affiliation(s)
- Riley E Scalzo
- Department of Pharmacy, Massachusetts General Hospital, Boston, Massachusetts
| | - Scott L Sanoff
- Department of Medicine, Duke University Hospital, Durham, North Carolina
| | - Aparna S Rege
- Department of Surgery, Duke University Hospital, Durham, North Carolina
| | - Jean Kwun
- Department of Surgery, Duke University Hospital, Durham, North Carolina
| | - Stuart J Knechtle
- Department of Surgery, Duke University Hospital, Durham, North Carolina
| | - Laura Barisoni
- Department of Pathology, Duke University Hospital, Durham, North Carolina
| | - Jennifer S Byrns
- Department of Pharmacy, Duke University Hospital, Durham, North Carolina.
| |
Collapse
|
26
|
Hill JA, Kiem ES, Bhatti A, Liu W, Keane-Candib J, Fitzpatrick KS, Boonyaratanakornkit J, Gardner RA, Green DJ, Maloney DG, Turtle CJ, Smith JM, Gimferrer I, Blosser CD, Jackson SW. Anti-HLA antibodies in recipients of CD19 versus BCMA-targeted CAR T-cell therapy. Am J Transplant 2023; 23:416-422. [PMID: 36748802 PMCID: PMC10266802 DOI: 10.1016/j.ajt.2022.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 10/05/2022] [Accepted: 11/06/2022] [Indexed: 01/15/2023]
Abstract
Antibodies against foreign human leukocyte antigen (HLA) molecules are barriers to successful organ transplantation. B cell-depleting treatments are used to reduce anti-HLA antibodies but have limited efficacy. We hypothesized that the primary source for anti-HLA antibodies is long-lived plasma cells, which are ineffectively targeted by B cell depletion. To study this, we screened for anti-HLA antibodies in a prospectively enrolled cohort of 49 patients who received chimeric antigen receptor T-cell therapy (CARTx), targeting naïve and memory B cells (CD19-targeted, n = 21) or plasma cells (BCMA-targeted, n = 28) for hematologic malignancies. Longitudinal samples were collected before and up to 1 year after CARTx. All individuals were in sustained remission. We identified 4 participants with anti-HLA antibodies before CD19-CARTx. Despite B cell depletion, anti-HLA antibodies and calculated panel reactive antibody scores were stable for 1 year after CD19-CARTx. Only 1 BCMA-CARTx recipient had pre-CARTx low-level anti-HLA antibodies, with no follow-up samples available. These data implicate CD19neg long-lived plasma cells as an important source for anti-HLA antibodies, a model supported by infrequent HLA sensitization in BCMA-CARTx subjects receiving previous plasma cell-targeted therapies. Thus, plasma cell-targeted therapies may be more effective against HLA antibodies, thereby enabling improved access to organ transplantation and rejection management.
Collapse
Affiliation(s)
- Joshua A Hill
- Departments of Medicine, University of Washington School of Medicine, Seattle, Washington, USA; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA.
| | - Erika S Kiem
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Atif Bhatti
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Winnie Liu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Jacob Keane-Candib
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Kristin S Fitzpatrick
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA; Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Jim Boonyaratanakornkit
- Departments of Medicine, University of Washington School of Medicine, Seattle, Washington, USA; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Rebecca A Gardner
- Seattle Children's Research Institute, Seattle, Washington, USA; Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
| | - Damian J Green
- Departments of Medicine, University of Washington School of Medicine, Seattle, Washington, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - David G Maloney
- Departments of Medicine, University of Washington School of Medicine, Seattle, Washington, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Cameron J Turtle
- Departments of Medicine, University of Washington School of Medicine, Seattle, Washington, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Jodi M Smith
- Seattle Children's Research Institute, Seattle, Washington, USA; Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
| | - Idoia Gimferrer
- Immunogenetics/HLA laboratory Bloodworks Northwest, Seattle, Washington, USA
| | - Christopher D Blosser
- Departments of Medicine, University of Washington School of Medicine, Seattle, Washington, USA; Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
| | - Shaun W Jackson
- Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA; Seattle Children's Research Institute, Seattle, Washington, USA; Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA.
| |
Collapse
|
27
|
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
|
28
|
Kikić Ž, Adam BA, Buxeda A, Lefaucheur C, Loupy A, Regele H, Cejka D, Haas M, Colvin RB, Mengel M. Quantitative scoring of progression in transplant glomerulopathy using digital pathology may be superior to Banff cg scoring. Kidney Int 2023; 103:365-377. [PMID: 36436680 DOI: 10.1016/j.kint.2022.10.024] [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: 04/22/2022] [Revised: 08/31/2022] [Accepted: 10/31/2022] [Indexed: 11/27/2022]
Abstract
Antibody-mediated rejection (ABMR) is a major cause of kidney allograft failure. Biopsy-based surrogate endpoints reflecting ABMR progression on sequential biopsies that predict long-term outcome offer the potential to make treatment trials for ABMR feasible. However, the Banff transplant glomerulopathy (TG) scoring system (chronic glomerular injury score [cg]) relies on relatively crude and arbitrary ordinal grades and has low inter-observer concordance that currently limits its usefulness as a surrogate endpoint for ABMR progression in clinical drug trials. Here, we describe and validate a novel quantitative method for quantifying progression of TG in ABMR. Using digital pathology in sequential biopsies from 75 patients at various stages of ABMR, we scored all capillaries in the most affected glomeruli for basement membrane duplication that were correlated with allograft function, outcome, Banff lesion scores, and gene expression. Our digital scoring reflected TG progression better than the categorical Banff cg score and correlated with Banff ABMR and chronicity lesions, but not transcript changes. In multivariate analysis, the delta change between biopsies with serum creatinine and mean percent duplicated glomerular basement membranes was significantly associated with graft loss. Neither the delta in any Banff lesion scores (including cg) nor in gene expression was associated with outcome. Receiver operating characteristic curve analysis showed that the digital pathology approach was superior to the conventional score for predicting graft failure. Thus, our digital pathology-based approach for scoring TG accurately assessed progression in TG. However, further validation as a potential surrogate endpoint in clinical trials for the treatment of ABMR is warranted.
Collapse
Affiliation(s)
- Željko Kikić
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna, Vienna, Austria; Department of Urology, Medical University Vienna, Vienna, Austria
| | - Benjamin A Adam
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Anna Buxeda
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | | | | | - Heinz Regele
- Institute of Clinical Pathology, Medical University Vienna, Vienna, Austria
| | - Daniel Cejka
- Division of Nephrology and Dialysis, Ordensklinikum Linz-Elisabethinen, Linz, Austria
| | - Mark Haas
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Robert B Colvin
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Mengel
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
29
|
Grimaldi V, Pagano M, Moccia G, Maiello C, De Rosa P, Napoli C. Novel insights in the clinical management of hyperimmune patients before and after transplantation. CURRENT RESEARCH IN IMMUNOLOGY 2023; 4:100056. [PMID: 36714552 PMCID: PMC9876744 DOI: 10.1016/j.crimmu.2023.100056] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 01/11/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Despite improvements in anti-Human Leucocyte Antigens antibody detection, identification, and characterization offer a better in peri-operative management techniques, antibodies remain a serious cause of morbidity and mortality for patients both before and after organ transplantation. Hyperimmune patients are disadvantaged by having to wait longer to receive an organ from a suitably matched donor. They could benefit from desensitization protocols in both pre- and post-transplantation period. Clinical studies are underway to highlight which best desensitization strategies could be assure the best outcome in both heart and kidney transplantation. Although most clinical evidence about desensitization strategies by using anti-CD20 monoclonal antibodies, proteasome inhibitors, anti-CD38 monoclonal antibodies, interleukin-6 blockade, cysteine protease and complement inhibitors, comes from kidney transplantation studies, many of the debated novel concepts can be easily applied to desensitization also in heart transplantation. Here, we discuss the candidates and recipients' management by using most common standard of care and novel therapeutics, desensitization endpoints, and strategies for future studies.
Collapse
Affiliation(s)
- Vincenzo Grimaldi
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology. Regional Reference Laboratory of Transplant Immunology (LIT) (EFI and ASHI Certifications). Department of Internal Medicine and Specialistics, University of Campania "L. Vanvitelli", Naples, Italy,Corresponding author.
| | - Martina Pagano
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology. Regional Reference Laboratory of Transplant Immunology (LIT) (EFI and ASHI Certifications). Department of Internal Medicine and Specialistics, University of Campania "L. Vanvitelli", Naples, Italy
| | - Giusi Moccia
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology. Regional Reference Laboratory of Transplant Immunology (LIT) (EFI and ASHI Certifications). Department of Internal Medicine and Specialistics, University of Campania "L. Vanvitelli", Naples, Italy
| | - Ciro Maiello
- Cardiac Transplantation Unit, Department of Cardiac Surgery and Transplantation, Ospedali dei Colli, Naples, Italy
| | - Paride De Rosa
- General Surgery and Transplantation Unit, "San Giovanni di Dio e Ruggi D'Aragona," University Hospital, Scuola Medica Salernitana, Salerno, Italy
| | - Claudio Napoli
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology. Regional Reference Laboratory of Transplant Immunology (LIT) (EFI and ASHI Certifications). Department of Internal Medicine and Specialistics, University of Campania "L. Vanvitelli", Naples, Italy,Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
30
|
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: 1.0] [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
|
31
|
Updated management for antibody-mediated rejection: opportunity to prolong kidney allograft survival. Curr Opin Nephrol Hypertens 2023; 32:13-19. [PMID: 36250450 DOI: 10.1097/mnh.0000000000000843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Antibody-mediated rejection (ABMR) is an important barrier to achieve long-term kidney allograft survival. Human leukocyte antibody (HLA)-incompatibility and ABO-incompatibility are the two main mechanisms of ABMR. Nevertheless, the advances in managing ABMR have changed the paradigm for kidney transplantation. This review aimed to emphasize the HLA-incompatibility and ABO-incompatibility kidney transplant and update the management of ABMR. RECENT FINDINGS HLA-incompatibility kidney transplantation is a strong risk factor for ABMR. Donor-specific antibody (DSA) is a surrogate biomarker that prevents long-term allograft survival. The standard treatment for ABMR has unfavorable results. New drugs that target the B cell are a promising approach to treat ABMR. In the past, ABO-incompatibility kidney donor was an absolute contraindication but now, it is widely accepted as an alternative organ resource. The advancement of ABO antibody removal and B-cell depletion therapy has been successfully developed. ABO isoagglutination remains the main biomarker for monitoring ABMR during the transplantation process. C4d staining without inflammation of the kidney allograft is the marker for the accommodation process. SUMMARY With the shortage of organ donors, transplant experts have expanded the organ resources and learned how to overcome the immunological barriers by using novel biomarkers and developing new treatments that support long-term graft survival.
Collapse
|
32
|
Giovannini D, Belbezier A, Baillet A, Bouillet L, Kawano M, Dumestre-Perard C, Clavarino G, Noble J, Pers JO, Sturm N, Huard B. Heterogeneity of antibody-secreting cells infiltrating autoimmune tissues. Front Immunol 2023; 14:1111366. [PMID: 36895558 PMCID: PMC9989216 DOI: 10.3389/fimmu.2023.1111366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/27/2023] [Indexed: 02/23/2023] Open
Abstract
The humoral response is frequently dysfunctioning in autoimmunity with a frequent rise in total serum immunoglobulins, among which are found autoantibodies that may be pathogenic by themselves and/or propagate the inflammatory reaction. The infiltration of autoimmune tissues by antibody-secreting cells (ASCs) constitutes another dysfunction. The known high dependency of ASCs on the microenvironment to survive combined to the high diversity of infiltrated tissues implies that ASCs must adapt. Some tissues even within a single clinical autoimmune entity are devoid of infiltration. The latter means that either the tissue is not permissive or ASCs fail to adapt. The origin of infiltrated ASCs is also variable. Indeed, ASCs may be commonly generated in the secondary lymphoid organ draining the autoimmune tissue, and home at the inflammation site under the guidance of specific chemokines. Alternatively, ASCs may be generated locally, when ectopic germinal centers are formed in the autoimmune tissue. Alloimmune tissues with the example of kidney transplantation will also be discussed own to their high similarity with autoimmune tissues. It should also be noted that antibody production is not the only function of ASCs, since cells with regulatory functions have also been described. This article will review all the phenotypic variations indicative of tissue adaptation described so for at the level of ASC-infiltrating auto/alloimmune tissues. The aim is to potentially define tissue-specific molecular targets in ASCs to improve the specificity of future autoimmune treatments.
Collapse
Affiliation(s)
- Diane Giovannini
- Department of Pathology, Grenoble University Hospital, Grenoble, France.,Translational Research in Autoimmunity and Inflammation Group (TRAIG), Translational Innovation in Medicine and Complexity (TIMC), University Grenoble-Alpes, CNRS Unité mixte de recherche (UMR) 5525, Grenoble, France
| | - Aude Belbezier
- Translational Research in Autoimmunity and Inflammation Group (TRAIG), Translational Innovation in Medicine and Complexity (TIMC), University Grenoble-Alpes, CNRS Unité mixte de recherche (UMR) 5525, Grenoble, France.,Department of Internal Medicine, Grenoble University Hospital, Grenoble, France
| | - Athan Baillet
- Translational Research in Autoimmunity and Inflammation Group (TRAIG), Translational Innovation in Medicine and Complexity (TIMC), University Grenoble-Alpes, CNRS Unité mixte de recherche (UMR) 5525, Grenoble, France.,Department of Rheumatology, Grenoble University Hospital, Grenoble, France
| | - Laurence Bouillet
- Translational Research in Autoimmunity and Inflammation Group (TRAIG), Translational Innovation in Medicine and Complexity (TIMC), University Grenoble-Alpes, CNRS Unité mixte de recherche (UMR) 5525, Grenoble, France.,Department of Internal Medicine, Grenoble University Hospital, Grenoble, France
| | - Mitsuhiro Kawano
- Department of Rheumatology, Kanazawa University Hospital, Kanazawa, Japan
| | | | | | - Johan Noble
- Department of Nephrology, Grenoble University Hospital, Grenoble, France
| | - Jacques-Olivier Pers
- B Lymphocytes, Autoimmunity and Immunotherapies, Brest University, INSERM, UMR1227, Brest, France.,Odontology Unit, Brest University Hospital, Brest, France
| | - Nathalie Sturm
- Department of Pathology, Grenoble University Hospital, Grenoble, France.,Translational Research in Autoimmunity and Inflammation Group (TRAIG), Translational Innovation in Medicine and Complexity (TIMC), University Grenoble-Alpes, CNRS Unité mixte de recherche (UMR) 5525, Grenoble, France
| | - Bertrand Huard
- Translational Research in Autoimmunity and Inflammation Group (TRAIG), Translational Innovation in Medicine and Complexity (TIMC), University Grenoble-Alpes, CNRS Unité mixte de recherche (UMR) 5525, Grenoble, France
| |
Collapse
|
33
|
Abstract
Antibody-mediated rejection (AMR) has a strongly negative impact on long-term renal allograft survival. Currently, no recognized effective treatments are available, especially for chronic antibody-mediated rejection (CAMR). Donor-specific antibodies (DSAs) secreted by long-lived plasma cells and memory B cells are acknowledged as biomarkers of AMR. Nevertheless, it may be too late for the DSA routine examination production since DSAs may have binded to graft vascular endothelial cells through complement-dependent or complement-independent pathways. Therefore, methods to effectively monitor memory B cells and long-lived plasma cells and subsequently prevent DSA production are key to reducing the adverse effects of AMR. Therefore, this review mainly summarizes the production pathways of memory B cells and long-lived plasma cells and provides suggestions for the prevention of AMR after transplantation.
Collapse
Affiliation(s)
- Wenlong Yue
- Kidney Transplantation Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jia Liu
- Dietetics Teaching and Research Section, Henan Medical College, Xinzheng, People's Republic of China
| | - Xiaohu Li
- Kidney Transplantation Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Luman Wang
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jinfeng Li
- Kidney Transplantation Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
34
|
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: 0.3] [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
|
35
|
Li X, Zhu H, Sui T, Zhao X, Deng Q. A Case of Daratumumab-Induced Significant Decrease in Donor-Specific HLA Antibodies and Remission Induction Before Haploidentical Stem Cell Transplantation in a Refractory B-ALL Patient. Cell Transplant 2022; 31:9636897221132502. [PMID: 36278404 PMCID: PMC9597015 DOI: 10.1177/09636897221132502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective: To explore the method of eliminating donor-specific anti-HLA antibodies (DSA)
in haploidentical stem cell transplantation (haplo-SCT). Methods: We present a refractory B-cell acute lymphoblastic leukemia (ALL) patient who
had strongly positive DSA, but had no human leukocyte antigen–matched donor.
Although CD38 expression on leukemia cells was negative, daratumumab
combined with etoposide and venetoclax therapy was chosen for her. Results: She achieved a significant decrease in DSA levels and complete remission on
the combination therapy with daratumumab. She then received a haplo-SCT from
a daughter as a donor and had a successful engraftment of donor stem cell.
In haplo-SCT, strongly positive DSA levels, directed against donor HLA
antigens, could be significantly reduced by daratumumab therapy before
transplantation and successfully bridge subsequent haplo-SCT. Conclusion: Although CD38 expression is negative in leukemia cells, refractory B-ALL
patients may still benefit from combination therapy with daratumumab. We
need further clinical observation.
Collapse
Affiliation(s)
- Xin Li
- Department of Hematology, Tianjin First
Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Haibo Zhu
- Department of Hematology, Tianjin First
Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Tao Sui
- Department of Hematology, Tianjin First
Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Xingli Zhao
- Departments of Oncology and Hematology,
Tianjin People’s Hospital, School of Medicine, Nankai University, Tianjin,
China
| | - Qi Deng
- Department of Hematology, Tianjin First
Central Hospital, School of Medicine, Nankai University, Tianjin, China,Qi Deng, Department of Hematology, Tianjin
First Central Hospital, School of Medicine, Nankai University, No. 24, Fukang
Road, Tianjin 300192, China.
| |
Collapse
|
36
|
Ho CS, Putnam KR, Peiter CR, Herczyk WF, Gerlach JA, Lu Y, Campagnaro EL, Woodside KJ, Cusick MF. Daratumumab Interferes with Allogeneic Crossmatch Impacting Immunological Assessment in Solid Organ Transplantation. J Clin Med 2022; 11:jcm11206059. [PMID: 36294380 PMCID: PMC9605360 DOI: 10.3390/jcm11206059] [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: 09/11/2022] [Revised: 10/03/2022] [Accepted: 10/12/2022] [Indexed: 11/07/2022] Open
Abstract
We report the first case of Daratumumab interference of allogeneic crossmatch tests repeatedly causing aberrant false-positive results, which inadvertently delayed transplant for a waitlisted renal patient with multiple myeloma. Daratumumab is an IgG1κ human monoclonal antibody commonly used to treat multiple myeloma, characterized by cancerous plasma cells and often leads to renal failure requiring kidney transplant, by depleting CD38-expressing plasma cells. In this case study, the patient had end-stage renal disease secondary to multiple myeloma and was continuously receiving Daratumumab infusions. The patient did not have any detectable antibodies to human leukocyte antigens but repeatedly had unexpected positive crossmatch by the flow cytometry-based method with 26 of the 27 potential deceased organ donors, implying donor-recipient immunological incompatibility. However, further review and analysis suggested that the positive crossmatches were likely false-positive as a result of interference from Daratumumab binding to donor cell surface CD38 as opposed to the presence of donor-specific antibodies. The observed intensity of the false-positive crossmatches was also highly variable, potentially due to donor- and/or cell-dependent expression of CD38. The variability of CD38 expression was, therefore, for the first time, characterized on the T and B cells isolated from various tissues and peripheral blood of 78 individuals. Overall, T cells were found to have a lower CD38 expression profile than the B cells, and no significant difference was observed between deceased and living individuals. Finally, we show that a simple cell treatment by dithiothreitol can effectively mitigate Daratumumab interference thus preserving the utility of pre-transplant crossmatch in multiple myeloma patients awaiting kidney transplant.
Collapse
Affiliation(s)
- Chak-Sum Ho
- Gift of Hope Organ & Tissue Donor Network, Itasca, IL 60143, USA
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
- Gift of Life Michigan, Ann Arbor, MI 48108, USA
- Correspondence: (C.-S.H.); (M.F.C.)
| | | | | | | | - John A. Gerlach
- Gift of Life Michigan, Ann Arbor, MI 48108, USA
- Biomedical Laboratory Diagnostics Program, College of Natural Science, Michigan State University, East Lansing, MI 48824, USA
| | - Yee Lu
- Division of Nephrology, Department of Internal Medicine, University of Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Erica L. Campagnaro
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Kenneth J. Woodside
- Division of Nephrology, Department of Internal Medicine, University of Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Matthew F. Cusick
- Department of Pathology, University of Michigan Medicine, Ann Arbor, MI 48109, USA
- Correspondence: (C.-S.H.); (M.F.C.)
| |
Collapse
|
37
|
Chong AS, Habal MV. From bench to bedside: reversing established antibody responses and desensitization. Curr Opin Organ Transplant 2022; 27:376-384. [PMID: 35950890 PMCID: PMC9474614 DOI: 10.1097/mot.0000000000001009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Basic transplant immunology has primarily focused on the definition of mechanisms, but an often-stated aspirational goal is to translate basic mechanistic research into future therapy. Pretransplant donor-specific antibodies (DSA) mediate hyperacute as well as early antibody-mediated rejection (AMR), whereas DSA developing late posttransplantation may additionally mediate chronic rejection. Although contemporary immunosuppression effectively prevents early cellular rejection after transplant in nonsensitized patients, it is less effective at controlling preexisting HLA antibody responses or reversing DSA once established, thus underscoring a need for better therapies. RECENT FINDINGS We here review the development of a bench-to-bedside approach involving transient proteasome inhibition to deplete plasma cells, combined with maintenance co-stimulation blockade, with CTLA-4Ig or belatacept, to prevent the generation of new antibody-secreting cells (ASCs). SUMMARY This review discusses how this treatment regimen, which was rationally designed and validated to reverse established DSA responses in mouse models, translated into reversing active AMR in the clinic, as well as desensitizing highly sensitized patients on the transplant waitlist.
Collapse
Affiliation(s)
- Anita S. Chong
- Department of Surgery, The University of Chicago, Chicago, Illinois, USA
| | - Marlena V. Habal
- Department of Medicine, Columbia University College of Medicine, New York, NY, USA
| |
Collapse
|
38
|
Abstract
PURPOSE OF THE REVIEW Antibody-mediated rejection (AMR) is the leading cause of kidney graft loss. Very few treatment options are available to the clinician to counter this disease process. In this review we describe the available therapeutics and the novel approaches that are being currently developed. RECENT FINDINGS AMR treatment requires a multidrug approach. Imlifidase, a new immunoglobulin G cleaving agent, may prove to be the perfect replacement of apheresis. New complement blockers other than eculizumab are in development in order to block acute kidney damage in the delicate phase following antibody removal. Plasma cell depletion is being explored in chronic AMR: studies are in progress with daratumumab and felzartamab. Interleukin 6 inhibition is generating enthusiasm in the chronic setting with preliminary encouraging results. SUMMARY In acute AMR, the clinicians will have to remove the antibodies, avoid rebound and block specific damage effectors. In chronic AMR they will need to reduce the inflammatory response induced by donor specific antibodies. New drugs are available and transplant physicians are starting to develop effective multidrug strategies to counter the complex disease mechanisms. Safety of these drugs needs to be further explored especially when used together with other potent immunosuppressive drugs.
Collapse
Affiliation(s)
- Paolo Malvezzi
- University Grenoble Alpes - CHU Grenoble Alpes - Service de Néphrologie, Dialyse, Aphérèses et Transplantation, Grenoble, France
| |
Collapse
|
39
|
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: 1.7] [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
|
40
|
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
|
41
|
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
|
42
|
Targeting CD38 in Neoplasms and Non-Cancer Diseases. Cancers (Basel) 2022; 14:cancers14174169. [PMID: 36077708 PMCID: PMC9454480 DOI: 10.3390/cancers14174169] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/21/2022] [Accepted: 08/25/2022] [Indexed: 01/12/2023] Open
Abstract
Simple Summary CD38 remains an interesting target for anticancer therapy. Its relatively high abundance in neoplasms and crucial impact on NAD+/cADPR metabolism and the activity of T cells allows for changing the immune response in autoimmune diseases, neoplasms, and finally the induction of cell death. Antibody-dependent cell cytotoxicity is responsible for cell death induced by targeting the tumor with anti-CD38 antibodies, such as daratumumab. A wide range of laboratory experiments and clinical trials show an especially promising role of anti-CD38 therapy against multiple myeloma, NK cell lymphomas, and CD19- B-cell malignancies. More studies are required to include more diseases in the therapeutic protocols involving the modulation of CD38 activity. Abstract CD38 is a myeloid antigen present both on the cell membrane and in the intracellular compartment of the cell. Its occurrence is often enhanced in cancer cells, thus making it a potential target in anticancer therapy. Daratumumab and isatuximab already received FDA approval, and novel agents such as MOR202, TAK079 and TNB-738 undergo clinical trials. Also, novel therapeutics such as SAR442085 aim to outrank the older antibodies against CD38. Multiple myeloma and immunoglobulin light-chain amyloidosis may be effectively treated with anti-CD38 immunotherapy. Its role in other hematological malignancies is also important concerning both diagnostic process and potential treatment in the future. Aside from the hematological malignancies, CD38 remains a potential target in gastrointestinal, neurological and pulmonary system disorders. Due to the strong interaction of CD38 with TCR and CD16 on T cells, it may also serve as the biomarker in transplant rejection in renal transplant patients. Besides, CD38 finds its role outside oncology in systemic lupus erythematosus and collagen-induced arthritis. CD38 plays an important role in viral infections, including AIDS and COVID-19. Most of the undergoing clinical trials focus on the use of anti-CD38 antibodies in the therapy of multiple myeloma, CD19- B-cell malignancies, and NK cell lymphomas. This review focuses on targeting CD38 in cancer and non-cancerous diseases using antibodies, cell-based therapies and CD38 inhibitors. We also provide a summary of current clinical trials targeting CD38.
Collapse
|
43
|
Halloran PF, Madill‐Thomsen KS, Pon S, Sikosana MLN, Böhmig GA, Bromberg J, Einecke G, Eskandary F, Gupta G, Hidalgo LG, Myslak M, Viklicky O, Perkowska‐Ptasinska A. Molecular diagnosis of ABMR with or without donor-specific antibody in kidney transplant biopsies: Differences in timing and intensity but similar mechanisms and outcomes. Am J Transplant 2022; 22:1976-1991. [PMID: 35575435 PMCID: PMC9540308 DOI: 10.1111/ajt.17092] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We studied the clinical, histologic, and molecular features distinguishing DSA-negative from DSA-positive molecularly defined antibody-mediated rejection (mABMR). We analyzed mABMR biopsies with available DSA assessments from the INTERCOMEX study: 148 DSA-negative versus 248 DSA-positive, compared with 864 no rejection (excluding TCMR and Mixed). DSA-positivity varied with mABMR stage: early-stage (EABMR) 56%; fully developed (FABMR) 70%; and late-stage (LABMR) 58%. DSA-negative patients with mABMR were usually sensitized, 60% being HLA antibody-positive. Compared with DSA-positive mABMR, DSA-negative mABMR was more often C4d-negative; earlier by 1.5 years (average 2.4 vs. 3.9 years); and had lower ABMR activity and earlier stage in molecular and histology features. However, the top ABMR-associated transcripts were identical in DSA-negative versus DSA-positive mABMR, for example, NK-associated (e.g., KLRD1 and GZMB) and IFNG-inducible (e.g., PLA1A). Genome-wide class comparison between DSA-negative and DSA-positive mABMR showed no significant differences in transcript expression except those related to lower intensity and earlier time of DSA-negative ABMR. Three-year graft loss in DSA-negative mABMR was the same as DSA-positive mABMR, even after adjusting for ABMR stage. Thus, compared with DSA-positive mABMR, DSA-negative mABMR is on average earlier, less active, and more often C4d-negative but has similar graft loss, and genome-wide analysis suggests that it involves the same mechanisms. SUMMARY SENTENCE: In 398 kidney transplant biopsies with molecular antibody-mediated rejection, the 150 DSA-negative cases are earlier, less intense, and mostly C4d-negative, but use identical molecular mechanisms and have the same risk of graft loss as the 248 DSA-positive cases.
Collapse
Affiliation(s)
- Philip F. Halloran
- Alberta Transplant Applied Genomics CentreEdmontonAlbertaCanada,Department of Medicine, Division of Nephrology and Transplant ImmunologyUniversity of AlbertaEdmontonAlbertaCanada
| | | | - Shane Pon
- Alberta Transplant Applied Genomics CentreEdmontonAlbertaCanada
| | | | - Georg A. Böhmig
- Division of Nephrology and Dialysis, Department of Medicine IIIMedical University of ViennaViennaAustria
| | | | - Gunilla Einecke
- Department of NephrologyHannover Medical SchoolHannoverGermany
| | - Farsad Eskandary
- Division of Nephrology and Dialysis, Department of Medicine IIIMedical University of ViennaViennaAustria
| | - Gaurav Gupta
- Division of NephrologyVirginia Commonwealth UniversityRichmondVirginiaUSA
| | | | - Marek Myslak
- Department of Clinical Interventions, Department of Nephrology and Kidney Transplantation SPWSZ HospitalPomeranian Medical UniversitySzczecinPoland
| | - Ondrej Viklicky
- Department of Nephrology and Transplant CenterInstitute for Clinical and Experimental MedicinePragueCzech Republic
| | | | | |
Collapse
|
44
|
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: 3.3] [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
|
45
|
Anwar IJ, DeLaura IF, Gao Q, Ladowski J, Jackson AM, Kwun J, Knechtle SJ. Harnessing the B Cell Response in Kidney Transplantation - Current State and Future Directions. Front Immunol 2022; 13:903068. [PMID: 35757745 PMCID: PMC9223638 DOI: 10.3389/fimmu.2022.903068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 04/25/2022] [Indexed: 01/21/2023] Open
Abstract
Despite dramatic improvement in kidney transplantation outcomes over the last decades due to advent of modern immunosuppressive agents, long-term outcomes remain poor. Antibody-mediated rejection (ABMR), a B cell driven process, accounts for the majority of chronic graft failures. There are currently no FDA-approved regimens for ABMR; however, several clinical trials are currently on-going. In this review, we present current mechanisms of B cell response in kidney transplantation, the clinical impact of sensitization and ABMR, the B cell response under current immunosuppressive regimens, and ongoing clinical trials for ABMR and desensitization treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Stuart J. Knechtle
- Duke Transplant Center, Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
46
|
Fenton M, Shaw K, Murchan H, Duignan S, Dunne E, McMahon CJ. Daratumumab Provides Transient Response Of Antibody Mediated Rejection Post Paediatric Orthotopic Heart Transplantation. J Heart Lung Transplant 2022; 41:1529-1530. [DOI: 10.1016/j.healun.2022.06.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/13/2022] [Accepted: 06/20/2022] [Indexed: 11/29/2022] Open
|
47
|
Benoit SW, Khandelwal P, Grimley MS. A case of treatment-resistant membranous nephropathy associated with graft versus host disease successfully treated with daratumumab. Pediatr Transplant 2022; 26:e14263. [PMID: 35249254 DOI: 10.1111/petr.14263] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 01/25/2022] [Accepted: 02/17/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Membranous nephropathy (MN) is the most common cause of glomerulopathy after hematopoietic cell transplantation (HCT), most often occurring in the setting of graft versus host disease (GVHD). Twenty percent of patients will fail to respond to standard therapy and may progress to end stage renal disease. Here we present the case of a pediatric patient who developed chronic oral GVHD more than one-year post-HCT, who subsequently developed nephrotic syndrome (anasarca, nephrotic range proteinuria, hypoalbuminemia) and had a renal biopsy consistent with MN. Treated with ibrutinib for her GVHD, and steroids, tacrolimus, and rituximab for her MN, she failed to achieve even partial remission of her kidney disease after 8 months. Due to steroid toxicity and 0% CD19 cells on lymphocyte subpopulation flow cytometry, the decision was made to trial plasma cell depletion therapy with daratumumab. METHOD She received three doses of daratumumab at weeks 1, 4, and 17. RESULTS Her nephrotic syndrome resolved and her serum albumin was greater than 3.0 gm/dl by week 10. She was weaned off of both steroids and tacrolimus by week 16, at which time she had near-complete remission of her renal disease. CONCLUSION Daratumumab may be an important, novel therapeutic option for post-HCT MN patients who are not responsive to standard therapies.
Collapse
Affiliation(s)
- Stefanie W Benoit
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Division of Bone Marrow Transplantation and Immunodeficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Pooja Khandelwal
- Division of Bone Marrow Transplantation and Immunodeficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Michael S Grimley
- Division of Bone Marrow Transplantation and Immunodeficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
48
|
Aghanejad A, Bonab SF, Sepehri M, Haghighi FS, Tarighatnia A, Kreiter C, Nader ND, Tohidkia MR. A review on targeting tumor microenvironment: The main paradigm shift in the mAb-based immunotherapy of solid tumors. Int J Biol Macromol 2022; 207:592-610. [PMID: 35296439 DOI: 10.1016/j.ijbiomac.2022.03.057] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 12/11/2022]
Abstract
Monoclonal antibodies (mAbs) as biological macromolecules have been remarked the large and growing pipline of the pharmaceutical market and also the most promising tool in modern medicine for cancer therapy. These therapeutic entities, which consist of whole mAbs, armed mAbs (i.e., antibody-toxin conjugates, antibody-drug conjugates, and antibody-radionuclide conjugates), and antibody fragments, mostly target tumor cells. However, due to intrinsic heterogeneity of cancer diseases, tumor cells targeting mAb have been encountered with difficulties in their unpredictable efficacy as well as variability in remission and durable clinical benefits among cancer patients. To address these pitfalls, the area has undergone two major evolutions with the intent of minimizing anti-drug responses and addressing limitations experienced with tumor cell-targeted therapies. As a novel hallmark of cancer, the tumor microenvironment (TME) is becoming the great importance of attention to develop innovative strategies based on therapeutic mAbs. Here, we underscore innovative strategies targeting TME by mAbs which destroy tumor cells indirectly through targeting vasculature system (e.g., anti-angiogenesis), immune system modulation (i.e., stimulation, suppression, and depletion), the targeting and blocking of stroma-based growth signals (e.g., cancer-associated fibroblasts), and targeting cancer stem cells, as well as, their effector mechanisms, clinical uses, and relevant mechanisms of resistance.
Collapse
Affiliation(s)
- Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samad Farashi Bonab
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Sepehri
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Sadat Haghighi
- Yazd Diabetes Research Center, Shahid Sadoghi University of Medical Sciences, Yazd, Iran
| | - Ali Tarighatnia
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Christopher Kreiter
- Department of Anesthesiology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| | - Nader D Nader
- Department of Anesthesiology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| | - Mohammad Reza Tohidkia
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
49
|
Mrak D, Bonelli M, Radner H. Neuropsychiatric Systemic Lupus Erythematosus: a remaining challenge. Curr Pharm Des 2022; 28:881-891. [PMID: 35549864 DOI: 10.2174/1381612828666220512102824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 04/13/2022] [Indexed: 11/22/2022]
Abstract
Systemic Lupus Erythematosus (SLE) is an autoimmune disease, which affects a wide range of organs with variable clinical features. Involvement of the nervous system is a challenging and multifaceted manifestation of the disease, presenting with a broad range of symptoms. Neuropsychiatric lupus (NPSLE) encompasses seven syndromes of the peripheral and 12 of the central nervous system, associated with a high disease burden. Despite advances in the management of SLE, NP manifestations still pose a challenge to clinicians. First, diagnosis and attribution to SLE is difficult due to the lack of specific biomarkers or imaging modalities. Second, therapeutic options are limited, and evidence is mainly based on case reports and expert consensus, as clinical trials are sparse. Moreover, no validated outcome measure on disease activity exists. Current recommendations for treatment include supportive as well as immunosuppressive medication, depending on the type and severity of manifestations. As NPSLE manifestations are increasingly recognized, a broader spectrum of therapeutic options can be expected.
Collapse
Affiliation(s)
- Daniel Mrak
- Medical University of Vienna, Vienna, Austria
| | - Michael Bonelli
- Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| | - Helga Radner
- Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
50
|
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: 2.3] [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
|