1
|
Del Bello A, Treiner E. Immune Checkpoints in Solid Organ Transplantation. BIOLOGY 2023; 12:1358. [PMID: 37887068 PMCID: PMC10604300 DOI: 10.3390/biology12101358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023]
Abstract
Allogenic graft acceptance is only achieved by life-long immunosuppression, which comes at the cost of significant toxicity. Clinicians face the challenge of adapting the patients' treatments over long periods to lower the risks associated with these toxicities, permanently leveraging the risk of excessive versus insufficient immunosuppression. A major goal and challenge in the field of solid organ transplantation (SOT) is to attain a state of stable immune tolerance specifically towards the grafted organ. The immune system is equipped with a set of inhibitory co-receptors known as immune checkpoints (ICs), which physiologically regulate numerous effector functions. Insufficient regulation through these ICs can lead to autoimmunity and/or immune-mediated toxicity, while excessive expression of ICs induces stable hypo-responsiveness, especially in T cells, a state sometimes referred to as exhaustion. IC blockade has emerged in the last decade as a powerful therapeutic tool against cancer. The opposite action, i.e., subverting IC for the benefit of establishing a state of specific hypo-responsiveness against auto- or allo-antigens, is still in its infancy. In this review, we will summarize the available literature on the role of ICs in SOT and the relevance of ICs with graft acceptance. We will also discuss the possible influence of current immunosuppressive medications on IC functions.
Collapse
Affiliation(s)
- Arnaud Del Bello
- Department of Nephrology, University Hospital of Toulouse, 31400 Toulouse, France
- Metabolic and Cardiovascular Research Institute (I2MC), Inserm UMR1297, CEDEX 4, 31432 Toulouse, France
- Faculty of Medicine, University Toulouse III Paul Sabatier, 31062 Toulouse, France
| | - Emmanuel Treiner
- Faculty of Medicine, University Toulouse III Paul Sabatier, 31062 Toulouse, France
- Laboratory of Immunology, University Hospital of Toulouse, 31300 Toulouse, France
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Inserm UMR1291, 31024 Toulouse, France
| |
Collapse
|
2
|
Sattler A, Schrezenmeier E, Weber UA, Potekhin A, Bachmann F, Straub-Hohenbleicher H, Budde K, Storz E, Proß V, Bergmann Y, Thole LM, Tizian C, Hölsken O, Diefenbach A, Schrezenmeier H, Jahrsdörfer B, Zemojtel T, Jechow K, Conrad C, Lukassen S, Stauch D, Lachmann N, Choi M, Halleck F, Kotsch K. Impaired humoral and cellular immunity after SARS-CoV-2 BNT162b2 (tozinameran) prime-boost vaccination in kidney transplant recipients. J Clin Invest 2021; 131:150175. [PMID: 34101623 PMCID: PMC8279581 DOI: 10.1172/jci150175] [Citation(s) in RCA: 207] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/03/2021] [Indexed: 12/15/2022] Open
Abstract
Novel mRNA-based vaccines have been proven to be powerful tools in combating the global pandemic caused by SARS-CoV-2, with BNT162b2 (trade name: Comirnaty) efficiently protecting individuals from COVID-19 across a broad age range. Still, it remains largely unknown how renal insufficiency and immunosuppressive medication affect development of vaccine-induced immunity. We therefore comprehensively analyzed humoral and cellular responses in kidney transplant recipients after the standard second vaccination dose. As opposed to all healthy vaccinees and the majority of hemodialysis patients, only 4 of 39 and 1 of 39 transplanted individuals showed IgA and IgG seroconversion at day 8 ± 1 after booster immunization, with minor changes until day 23 ± 5, respectively. Although most transplanted patients mounted spike-specific T helper cell responses, frequencies were significantly reduced compared with those in controls and dialysis patients and this was accompanied by a broad impairment in effector cytokine production, memory differentiation, and activation-related signatures. Spike-specific CD8+ T cell responses were less abundant than their CD4+ counterparts in healthy controls and hemodialysis patients and almost undetectable in transplant patients. Promotion of anti-HLA antibodies or acute rejection was not detected after vaccination. In summary, our data strongly suggest revised vaccination approaches in immunosuppressed patients, including individual immune monitoring for protection of this vulnerable group at risk of developing severe COVID-19.
Collapse
Affiliation(s)
| | - Eva Schrezenmeier
- Department of Nephrology and Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Ulrike A. Weber
- Department of Nephrology and Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Alexander Potekhin
- Department of Nephrology and Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- MVZ Diaverum Neubrandenburg, Neubrandenburg, Germany
| | - Friederike Bachmann
- Department of Nephrology and Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Henriette Straub-Hohenbleicher
- Department of Nephrology and Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Elena Storz
- Department for General and Visceral Surgery and
| | | | | | | | - Caroline Tizian
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany
| | - Oliver Hölsken
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany
- Heidelberg Bioscience International Graduate School, Heidelberg University, Heidelberg, Germany
| | - Andreas Diefenbach
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany
| | - Hubert Schrezenmeier
- Department of Transfusion Medicine and Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service, Baden-Württemberg—Hessen and University Hospital Ulm, Ulm University, Ulm, Germany
| | - Bernd Jahrsdörfer
- Department of Transfusion Medicine and Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service, Baden-Württemberg—Hessen and University Hospital Ulm, Ulm University, Ulm, Germany
| | | | | | | | | | - Diana Stauch
- HLA Laboratory, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany
| | - Nils Lachmann
- HLA Laboratory, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany
| | - Mira Choi
- Department of Nephrology and Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Fabian Halleck
- Department of Nephrology and Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | | |
Collapse
|
3
|
Kirchhof J, Wilde B, Schmidt J, Mülling N, Petrakova L, Brinkhoff A, Schedlowski M, Witzke O. Acute Versus Chronic Administration of Calcineurin-Inhibitors Differentially Affect T-Cell Function. Endocr Metab Immune Disord Drug Targets 2021; 21:1083-1089. [PMID: 32867664 DOI: 10.2174/1871530320999200831161710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/17/2020] [Accepted: 07/25/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Calcineurin-inhibitors (CNI) are used in renal transplant patients (RTX) to prevent rejection. CNI mainly suppress T-cell mediated immunity but very little is known about the impact of long-term treatment with CNI on T-cell function. OBJECTIVE We investigated the immunological effects of long-term CNI intake in RTX patients in comparison to short-term CNI administration in healthy controls (HC). METHODS Blood was drawn from 30 RTX patients with long-term CNI treatment. In addition, blood was sampled from HC with short-term CNI treatment (four dosages) before the first and 2 hours after the last CsA intake. T-cells were analyzed for cytokine production, proliferation, and CD25 expression. RESULTS Short-term CNI reduced T-cell derived IL-2 and IFNγ as well as T-cell proliferation in HC. IFNγ was not suppressed in patients with long-term CNI treatment. IL-2 production, CD25 expression, and T-cell proliferation were enhanced in long-term CNI patients. CONCLUSION Suppression of IFNγ/IL-2 and T-cell proliferation is weaker during long-term CNI treatment in patients compared to short-term treatment in healthy subjects. Enhanced CD25 expression may lower the threshold for T-cell activation during long-term CNI treatment.
Collapse
Affiliation(s)
- Julia Kirchhof
- Institute of Medical Psychology and Behavioral Immunology, University Hospital Essen, University of Duisburg- Essen, 45122 Essen, Germany
| | - Benjamin Wilde
- Department of Nephrology, University Hospital Essen, University of Duisburg- Essen, Germany
| | - Justine Schmidt
- Institute of Medical Psychology and Behavioral Immunology, University Hospital Essen, University of Duisburg- Essen, 45122 Essen, Germany
| | - Nils Mülling
- Department of Nephrology, University Hospital Essen, University of Duisburg- Essen, Germany
| | - Liubov Petrakova
- Institute of Medical Psychology and Behavioral Immunology, University Hospital Essen, University of Duisburg- Essen, 45122 Essen, Germany
| | - Alexandra Brinkhoff
- Department of Nephrology, University Hospital Essen, University of Duisburg- Essen, Germany
| | - Manfred Schedlowski
- Institute of Medical Psychology and Behavioral Immunology, University Hospital Essen, University of Duisburg- Essen, 45122 Essen, Germany
| | - Oliver Witzke
- Department of Infectious Diseases, West German Centre of Infectious Diseases, Universitätsmedizin Essen, University Duisburg-Essen, Duisburg, Germany
| |
Collapse
|
4
|
Abstract
Costimulation between T cells and antigen-presenting cells is essential for the regulation of an effective alloimmune response and is not targeted with the conventional immunosuppressive therapy after kidney transplantation. Costimulation blockade therapy with biologicals allows precise targeting of the immune response but without non-immune adverse events. Multiple costimulation blockade approaches have been developed that inhibit the alloimmune response in kidney transplant recipients with varying degrees of success. Belatacept, an immunosuppressive drug that selectively targets the CD28-CD80/CD86 pathway, is the only costimulation blockade therapy that is currently approved for kidney transplant recipients. In the last decade, belatacept therapy has been shown to be a promising therapy in subgroups of kidney transplant recipients; however, the widespread use of belatacept has been tempered by an increased risk of acute kidney transplant rejection. The purpose of this review is to provide an overview of the costimulation blockade therapies that are currently in use or being developed for kidney transplant indications.
Collapse
|
5
|
Marshall N, Hutchinson K, Marron TU, Aleynick M, Hammerich L, Upadhyay R, Svensson-Arvelund J, Brown BD, Merad M, Brody JD. Antitumor T-cell Homeostatic Activation Is Uncoupled from Homeostatic Inhibition by Checkpoint Blockade. Cancer Discov 2019; 9:1520-1537. [PMID: 31375522 DOI: 10.1158/2159-8290.cd-19-0391] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 06/18/2019] [Accepted: 07/16/2019] [Indexed: 12/21/2022]
Abstract
T-cell transfer into lymphodepleted recipients induces homeostatic activation and potentiates antitumor efficacy. In contrast to canonical T-cell receptor-induced activation, homeostatic activation yields a distinct phenotype and memory state whose regulatory mechanisms are poorly understood. Here, we show in patients and murine models that, following transfer into lymphodepleted bone marrow transplant (BMT) recipients, CD8+ T cells undergo activation but also simultaneous homeostatic inhibition manifested by upregulation of immune-checkpoint molecules and functional suppression. T cells transferred into BMT recipients were protected from homeostatic inhibition by PD-1/CTLA4 dual checkpoint blockade (dCB). This combination of dCB and BMT-"immunotransplant"-increased T-cell homeostatic activation and antitumor T-cell responses by an order of magnitude. Like homeostatic activation, homeostatic inhibition is IL7/IL15-dependent, revealing mechanistic coupling of these two processes. Marked similarity in ex vivo modulation of post-BMT T cells in mice and patients is promising for the clinical translation of immunotransplant (NCT03305445) and for addressing homeostatic inhibition in T-cell therapies. SIGNIFICANCE: For optimal anticancer effect, T-cell therapies including chimeric antigen receptor T-cell, tumor-infiltrating lymphocyte, and transgenic T-cell therapies require transfer into lymphodepleted recipients and homeostatic activation; however, concomitant homeostatic inhibition mitigates T-cell therapies' efficacy. Checkpoint blockade uncouples homeostatic inhibition from activation, amplifying T-cell responses. Conversely, tumors nonresponsive to checkpoint blockade or BMT are treatable with immunotransplant.See related commentary by Ansell, p. 1487.This article is highlighted in the In This Issue feature, p. 1469.
Collapse
Affiliation(s)
- Netonia Marshall
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine, Mount Sinai Hospital, New York, New York
| | - Keino Hutchinson
- Department of Pharmacological Sciences, Icahn School of Medicine, Mount Sinai Hospital, New York, New York
| | - Thomas U Marron
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine, Mount Sinai Hospital, New York, New York
| | - Mark Aleynick
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine, Mount Sinai Hospital, New York, New York
| | - Linda Hammerich
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine, Mount Sinai Hospital, New York, New York
| | - Ranjan Upadhyay
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine, Mount Sinai Hospital, New York, New York
| | - Judit Svensson-Arvelund
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine, Mount Sinai Hospital, New York, New York
| | - Brian D Brown
- Department of Genetics and Genomic Sciences, Icahn School of Medicine, Mount Sinai Hospital, New York, New York
| | - Miriam Merad
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine, Mount Sinai Hospital, New York, New York.,Department of Oncological Sciences, Icahn School of Medicine, Mount Sinai Hospital, New York, New York
| | - Joshua D Brody
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine, Mount Sinai Hospital, New York, New York.
| |
Collapse
|
6
|
van der Zwan M, Clahsen-Van Groningen MC, Roodnat JI, Bouvy AP, Slachmuylders CL, Weimar W, Baan CC, Hesselink DA, Kho MML. The Efficacy of Rabbit Anti-Thymocyte Globulin for Acute Kidney Transplant Rejection in Patients Using Calcineurin Inhibitor and Mycophenolate Mofetil-Based Immunosuppressive Therapy. Ann Transplant 2018; 23:577-590. [PMID: 30115901 PMCID: PMC6248318 DOI: 10.12659/aot.909646] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background T cell depleting antibody therapy with rabbit anti-thymocyte globulin (rATG) is the treatment of choice for glucocorticoid-resistant acute kidney allograft rejection (AR) and is used as first-line therapy in severe AR. Almost all studies investigating the effectiveness of rATG for this indication were conducted at the time when cyclosporine A and azathioprine were the standard of care. Here, the long-term outcome of rATG for AR in patients using the current standard immunosuppressive therapy (i.e., tacrolimus and mycophenolate mofetil) is described. Material/Methods Between 2002 to 2012, 108 patients were treated with rATG for AR. Data on kidney function in the year following rATG and long-term outcomes were collected. Results Overall survival after rATG was comparable to overall survival of all kidney transplantation patients (P=0.10). Serum creatinine 1 year after rATG was 179 μmol/L (interquartile range (IQR) 136–234 μmol/L) and was comparable to baseline serum creatinine (P=0.22). Early AR showed better allograft survival than late AR (P=0.0007). In addition, 1 year after AR, serum creatinine was lower in early AR (157 mol/L; IQR 131–203) compared to late AR (216 mol/L; IQR 165–269; P<0.05). The Banff grade of rejection, kidney function at the moment of rejection, and reason for rATG (severe or glucocorticoid resistant AR) did not influence the allograft survival. Conclusions Treatment of AR with rATG is effective in patients using current standard immunosuppressive therapy, even in patients with poor allograft function. Early identification of AR followed by T cell depleting treatment leads to better allograft outcomes.
Collapse
Affiliation(s)
- Marieke van der Zwan
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam Transplant Group, Rotterdam, Netherlands
| | - Marian C Clahsen-Van Groningen
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, Rotterdam Transplant Group, Rotterdam, Netherlands
| | - Joke I Roodnat
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam Transplant Group, Rotterdam, Netherlands
| | - Anne P Bouvy
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam Transplant Group, Rotterdam, Netherlands
| | - Casper L Slachmuylders
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam Transplant Group, Rotterdam, Netherlands
| | - Willem Weimar
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam Transplant Group, Rotterdam, Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam Transplant Group, Rotterdam, Netherlands
| | - Dennis A Hesselink
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam Transplant Group, Rotterdam, Netherlands
| | - Marcia M L Kho
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam Transplant Group, Rotterdam, Netherlands
| |
Collapse
|
7
|
Buermann A, Petkov S, Petersen B, Hein R, Lucas-Hahn A, Baars W, Brinkmann A, Niemann H, Schwinzer R. Pigs expressing the human inhibitory ligand PD-L1 (CD 274) provide a new source of xenogeneic cells and tissues with low immunogenic properties. Xenotransplantation 2018; 25:e12387. [DOI: 10.1111/xen.12387] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/27/2017] [Accepted: 01/12/2018] [Indexed: 01/01/2023]
Affiliation(s)
- Anna Buermann
- Transplant Laboratory; Department of General-, Visceral-, and Transplantation Surgery; Hannover Medical School; Hannover Germany
| | - Stoyan Petkov
- Institute of Farm Animal Genetics; Friedrich-Loeffler-Institut; Mariensee Germany
| | - Björn Petersen
- Institute of Farm Animal Genetics; Friedrich-Loeffler-Institut; Mariensee Germany
| | - Rabea Hein
- Transplant Laboratory; Department of General-, Visceral-, and Transplantation Surgery; Hannover Medical School; Hannover Germany
| | - Andrea Lucas-Hahn
- Institute of Farm Animal Genetics; Friedrich-Loeffler-Institut; Mariensee Germany
| | - Wiebke Baars
- Transplant Laboratory; Department of General-, Visceral-, and Transplantation Surgery; Hannover Medical School; Hannover Germany
| | - Antje Brinkmann
- Transplant Laboratory; Department of General-, Visceral-, and Transplantation Surgery; Hannover Medical School; Hannover Germany
| | - Heiner Niemann
- Institute of Farm Animal Genetics; Friedrich-Loeffler-Institut; Mariensee Germany
| | - Reinhard Schwinzer
- Transplant Laboratory; Department of General-, Visceral-, and Transplantation Surgery; Hannover Medical School; Hannover Germany
| |
Collapse
|
8
|
Kannegieter NM, Hesselink DA, Dieterich M, de Graav GN, Kraaijeveld R, Baan CC. Differential T Cell Signaling Pathway Activation by Tacrolimus and Belatacept after Kidney Transplantation: Post Hoc Analysis of a Randomised-Controlled Trial. Sci Rep 2017; 7:15135. [PMID: 29123208 PMCID: PMC5680251 DOI: 10.1038/s41598-017-15542-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/30/2017] [Indexed: 12/13/2022] Open
Abstract
Pharmacokinetic immunosuppressive drug monitoring poorly correlates with clinical outcomes after solid organ transplantation. A promising method for pharmacodynamic monitoring of tacrolimus (TAC) in T cell subsets of transplant recipients might be the measurement of (phosphorylated) p38MAPK, ERK1/2 and Akt (activated downstream of the T cell receptor) by phospho-specific flow cytometry. Here, blood samples from n = 40 kidney transplant recipients (treated with either TAC-based or belatacept (BELA)-based immunosuppressive drug therapy) were monitored before and throughout the first year after transplantation. After transplantation and in unstimulated samples, p-p38MAPK and p-Akt were inhibited in CD8+ T cells and p-ERK in CD4+ T cells but only in patients who received TAC-based therapy. After activation with PMA/ionomycin, p-p38MAPK and p-AKT were significantly inhibited in CD4+ and CD8+ T cells when TAC was given, compared to pre-transplantation. Eleven BELA-treated patients had a biopsy-proven acute rejection, which was associated with higher p-ERK levels in both CD4+ and CD8+ T cells compared to patients without rejection. In conclusion, phospho-specific flow cytometry is a promising tool to pharmacodynamically monitor TAC-based therapy. In contrast to TAC-based therapy, BELA-based immunosuppression does not inhibit key T cell activation pathways which may contribute to the high rejection incidence among BELA-treated transplant recipients.
Collapse
Affiliation(s)
- Nynke M Kannegieter
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - Dennis A Hesselink
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marjolein Dieterich
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Gretchen N de Graav
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rens Kraaijeveld
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
9
|
Alemtuzumab as Antirejection Therapy: T Cell Repopulation and Cytokine Responsiveness. Transplant Direct 2016; 2:e83. [PMID: 27500273 PMCID: PMC4946521 DOI: 10.1097/txd.0000000000000595] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 04/10/2016] [Indexed: 01/05/2023] Open
Abstract
Supplemental digital content is available in the text. Background Alemtuzumab induction therapy in kidney transplant patients results in T cell depletion followed by slow immune reconstitution of memory T cells with reduced immune functions. The kinetics and functional characteristics of T cell reconstitution when alemtuzumab is given during immune activation, ie, as antirejection therapy, are unknown. Methods Patients (n = 12) with glucocorticoid-resistant or severe vascular kidney transplant rejection were treated with alemtuzumab. Flow cytometric analysis was performed on whole blood to measure cell division by the marker Ki-67, and cytokine responsiveness by IL-2–mediated and IL-7–mediated phosphorylation of signal transducer and activator of transcription 5 of T cells before and during the first year after rejection therapy. Results At 1 year after alemtuzumab antirejection therapy, the total T cell population recovered to baseline level. Repopulation of CD4+ and CD8+ T cells was associated with increased percentages of Ki-67+ proliferating T cells (P < 0.05). In addition, both populations showed a phenotypic shift toward relatively more memory T cells (P < 0.01). At the functional level, IL-7 reactivity of CD4+ memory T cells was diminished, reflected by a decreased capacity to phosphorylate signal transducer and activator of transcription 5 during the first 6 months after alemtuzumab treatment (P < 0.05), whereas reactivity to IL-2 was preserved. CD8+ T cells were affected in terms of both IL-2 and IL-7 responses (both P < 0.05). After reconstitution, relatively more regulatory T cells were present, and a relatively high proportion of Ki-67+ T cells was observed. Conclusions Preliminary data from this small series suggest that alemtuzumab antirejection therapy induces homeostatic proliferation of memory and regulatory T cells with diminished responsiveness to the homeostatic cytokine IL-7. IL-2 responsiveness was affected in repopulated CD8+ T cells.
Collapse
|