1
|
Mittal R, McKenna K, Lemos JRN, Juneja S, Mittal M, Hirani K. Therapeutic potential of anti-thymocyte globulin in type 1 diabetes: A systematic review. PLoS One 2025; 20:e0323642. [PMID: 40359439 PMCID: PMC12074605 DOI: 10.1371/journal.pone.0323642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 04/13/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is an autoimmune condition characterized by the destruction of insulin-producing beta cells in the pancreas. Anti-Thymocyte Globulin (ATG) has emerged as a promising immunomodulatory therapy aimed at preserving beta-cell function and altering the disease course. This systematic review synthesizes current evidence from the clinical trials evaluating the efficacy and safety of low-dose ATG in individuals with T1D. METHODS We conducted a comprehensive literature search of electronic databases, including PubMed (MEDLINE), Science Direct, Scopus, EMBASE, and ClinicalTrials.gov, to identify studies investigating ATG in T1D in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) criteria. The Joanna Briggs Institute (JBI) Critical Appraisal Tools for randomized clinical trials and case-control studies were used to assess the quality and evaluate the risk of bias in the eligible studies. RESULTS The primary outcomes assessed were preservation of C-peptide levels, glycemic control, and adverse events. Results indicated that ATG showed potential in preserving beta-cell function and improving clinical outcomes in recent-onset T1D. However, the incidence of adverse events, such as cytokine release syndrome and lymphopenia, necessitated careful monitoring and management. CONCLUSION Low-dose ATG presents a promising therapeutic approach for modifying the progression of T1D. While early-phase trials demonstrate potential benefits in preserving beta-cell function, further large-scale, long-term studies are essential to establish optimal dosing regimens, long-term efficacy, and safety profiles. This review highlights the importance of continued research to fully elucidate the role of ATG in T1D management.
Collapse
Affiliation(s)
- Rahul Mittal
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Keelin McKenna
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, United States of America
| | - Joana R. N. Lemos
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Shreya Juneja
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Mannat Mittal
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Khemraj Hirani
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| |
Collapse
|
2
|
Iglesias M, Brennan DC, Larsen CP, Raimondi G. Targeting inflammation and immune activation to improve CTLA4-Ig-based modulation of transplant rejection. Front Immunol 2022; 13:926648. [PMID: 36119093 PMCID: PMC9478663 DOI: 10.3389/fimmu.2022.926648] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
For the last few decades, Calcineurin inhibitors (CNI)-based therapy has been the pillar of immunosuppression for prevention of organ transplant rejection. However, despite exerting effective control of acute rejection in the first year post-transplant, prolonged CNI use is associated with significant side effects and is not well suited for long term allograft survival. The implementation of Costimulation Blockade (CoB) therapies, based on the interruption of T cell costimulatory signals as strategy to control allo-responses, has proven potential for better management of transplant recipients compared to CNI-based therapies. The use of the biologic cytotoxic T-lymphocyte associated protein 4 (CTLA4)-Ig is the most successful approach to date in this arena. Following evaluation of the BENEFIT trials, Belatacept, a high-affinity version of CTLA4-Ig, has been FDA approved for use in kidney transplant recipients. Despite its benefits, the use of CTLA4-Ig as a monotherapy has proved to be insufficient to induce long-term allograft acceptance in several settings. Multiple studies have demonstrated that events that induce an acute inflammatory response with the consequent release of proinflammatory cytokines, and an abundance of allograft-reactive memory cells in the recipient, can prevent the induction of or break established immunomodulation induced with CoB regimens. This review highlights advances in our understanding of the factors and mechanisms that limit CoB regimens efficacy. We also discuss recent successes in experimentally designing complementary therapies that favor CTLA4-Ig effect, affording a better control of transplant rejection and supporting their clinical applicability.
Collapse
Affiliation(s)
- Marcos Iglesias
- Vascularized and Composite Allotransplantation (VCA) Laboratory, Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- *Correspondence: Giorgio Raimondi, ; Marcos Iglesias,
| | - Daniel C. Brennan
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Christian P. Larsen
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States
| | - Giorgio Raimondi
- Vascularized and Composite Allotransplantation (VCA) Laboratory, Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- *Correspondence: Giorgio Raimondi, ; Marcos Iglesias,
| |
Collapse
|
3
|
Usuelli V, Ben Nasr M, D'Addio F, Liu K, Vergani A, El Essawy B, Yang J, Assi E, Uehara M, Rossi C, Solini A, Capobianco A, Rigamonti E, Potena L, Venturini M, Sabatino M, Bottarelli L, Ammirati E, Frigerio M, Castillo‐Leon E, Maestroni A, Azzoni C, Loretelli C, Joe Seelam A, Tai AK, Pastore I, Becchi G, Corradi D, Visner GA, Zuccotti GV, Chau NB, Abdi R, Pezzolesi MG, Fiorina P. miR-21 antagonism reprograms macrophage metabolism and abrogates chronic allograft vasculopathy. Am J Transplant 2021; 21:3280-3295. [PMID: 33764625 PMCID: PMC8518036 DOI: 10.1111/ajt.16581] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/19/2021] [Accepted: 03/09/2021] [Indexed: 01/25/2023]
Abstract
Despite much progress in improving graft outcome during cardiac transplantation, chronic allograft vasculopathy (CAV) remains an impediment to long-term graft survival. MicroRNAs (miRNAs) emerged as regulators of the immune response. Here, we aimed to examine the miRNA network involved in CAV. miRNA profiling of heart samples obtained from a murine model of CAV and from cardiac-transplanted patients with CAV demonstrated that miR-21 was most significantly expressed and was primarily localized to macrophages. Interestingly, macrophage depletion with clodronate did not significantly prolong allograft survival in mice, while conditional deletion of miR-21 in macrophages or the use of a specific miR-21 antagomir resulted in indefinite cardiac allograft survival and abrogated CAV. The immunophenotype, secretome, ability to phagocytose, migration, and antigen presentation of macrophages were unaffected by miR-21 targeting, while macrophage metabolism was reprogrammed, with a shift toward oxidative phosphorylation in naïve macrophages and with an inhibition of glycolysis in pro-inflammatory macrophages. The aforementioned effects resulted in an increase in M2-like macrophages, which could be reverted by the addition of L-arginine. RNA-seq analysis confirmed alterations in arginase-associated pathways associated with miR-21 antagonism. In conclusion, miR-21 is overexpressed in murine and human CAV, and its targeting delays CAV onset by reprogramming macrophages metabolism.
Collapse
Affiliation(s)
- Vera Usuelli
- International Center for T1DPediatric Clinical Research Center “Romeo ed Enrica Invernizzi”Department of Biomedical and Clinical Science L. SaccoUniversita Degli Studi di MilanoMilanItaly
| | - Moufida Ben Nasr
- International Center for T1DPediatric Clinical Research Center “Romeo ed Enrica Invernizzi”Department of Biomedical and Clinical Science L. SaccoUniversita Degli Studi di MilanoMilanItaly,Nephrology DivisionBoston Children's HospitalHarvard Medical SchoolBostonMassachusetts
| | - Francesca D'Addio
- International Center for T1DPediatric Clinical Research Center “Romeo ed Enrica Invernizzi”Department of Biomedical and Clinical Science L. SaccoUniversita Degli Studi di MilanoMilanItaly
| | - Kaifeng Liu
- Division of Pulmonary and Respiratory DiseasesBoston Children's HospitalHarvard Medical SchoolBostonMassachusetts
| | - Andrea Vergani
- Nephrology DivisionBoston Children's HospitalHarvard Medical SchoolBostonMassachusetts
| | - Basset El Essawy
- Department of MedicineAl‐Azhar UniversityCairoEgypt,Renal DivisionTransplantation Research CenterBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusetts
| | - Jun Yang
- Institute of Organ TransplantationTongji Hospital and Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Emma Assi
- International Center for T1DPediatric Clinical Research Center “Romeo ed Enrica Invernizzi”Department of Biomedical and Clinical Science L. SaccoUniversita Degli Studi di MilanoMilanItaly
| | - Mayuko Uehara
- Renal DivisionTransplantation Research CenterBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusetts
| | - Chiara Rossi
- Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Anna Solini
- Department of SurgicalMedical, Molecular and Critical Area PathologyUniversity of PisaPisaItaly
| | - Annalisa Capobianco
- Division of Immunology, Transplantation and Infectious DiseaseSan Raffaele Scientific InstituteMilanItaly
| | - Elena Rigamonti
- Division of Immunology, Transplantation and Infectious DiseaseSan Raffaele Scientific InstituteMilanItaly
| | - Luciano Potena
- Heart Failure and Heart Transplant ProgramS. Orsola‐Malpighi HospitalAlma‐Mater University of BolognaBolognaItaly
| | | | - Mario Sabatino
- Department of Cardiothoracic, Transplantation and Vascular SurgeryS. Orsola‐Malpighi HospitalAlma Mater‐University of BolognaBolognaItaly
| | | | - Enrico Ammirati
- De Gasperis Cardio Center and Transplant CenterNiguarda HospitalMilanItaly
| | - Maria Frigerio
- De Gasperis Cardio Center and Transplant CenterNiguarda HospitalMilanItaly
| | - Eduardo Castillo‐Leon
- Nephrology DivisionBoston Children's HospitalHarvard Medical SchoolBostonMassachusetts
| | - Anna Maestroni
- International Center for T1DPediatric Clinical Research Center “Romeo ed Enrica Invernizzi”Department of Biomedical and Clinical Science L. SaccoUniversita Degli Studi di MilanoMilanItaly
| | - Cinzia Azzoni
- Department of Medicine and SurgeryUniversity of ParmaParmaItaly
| | - Cristian Loretelli
- International Center for T1DPediatric Clinical Research Center “Romeo ed Enrica Invernizzi”Department of Biomedical and Clinical Science L. SaccoUniversita Degli Studi di MilanoMilanItaly
| | - Andy Joe Seelam
- International Center for T1DPediatric Clinical Research Center “Romeo ed Enrica Invernizzi”Department of Biomedical and Clinical Science L. SaccoUniversita Degli Studi di MilanoMilanItaly
| | - Albert K. Tai
- Tufts University Core Facility (TUCF) Genomics CoreTufts University School of MedicineBostonMassachusetts
| | - Ida Pastore
- Division of EndocrinologyASST Fatebenefratelli‐SaccoMilanItaly
| | | | | | - Gary A. Visner
- Division of Pulmonary and Respiratory DiseasesBoston Children's HospitalHarvard Medical SchoolBostonMassachusetts
| | - Gian V. Zuccotti
- International Center for T1DPediatric Clinical Research Center “Romeo ed Enrica Invernizzi”Department of Biomedical and Clinical Science L. SaccoUniversita Degli Studi di MilanoMilanItaly,Department of PediatricsBuzzi Children's HospitalMilanItaly
| | | | - Reza Abdi
- Renal DivisionTransplantation Research CenterBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusetts
| | - Marcus G. Pezzolesi
- Division of Nephrology and Hypertension, Diabetes and Metabolism CenterUniversity of UtahSalt Lake CityUtah
| | - Paolo Fiorina
- International Center for T1DPediatric Clinical Research Center “Romeo ed Enrica Invernizzi”Department of Biomedical and Clinical Science L. SaccoUniversita Degli Studi di MilanoMilanItaly,Nephrology DivisionBoston Children's HospitalHarvard Medical SchoolBostonMassachusetts,Division of EndocrinologyASST Fatebenefratelli‐SaccoMilanItaly
| |
Collapse
|
4
|
Tomita Y, Iwadoh K, Ogawa Y, Miki K, Kato Y, Kai K, Sannomiya A, Koyama I, Kitajima K, Nakajima I, Fuchinoue S. Single fixed low-dose rituximab as induction therapy suppresses de novo donor-specific anti-HLA antibody production in ABO compatible living kidney transplant recipients. PLoS One 2019; 14:e0224203. [PMID: 31644555 PMCID: PMC6808551 DOI: 10.1371/journal.pone.0224203] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 10/08/2019] [Indexed: 01/09/2023] Open
Abstract
This study was conducted to evaluate de novo donor-specific anti-human leukocyte antigen (HLA) antibody (dnDSA) production leading to antibody-mediated rejection (ABMR) after rituximab induction in non-sensitized ABO-compatible living kidney transplantation (ABO-CLKTx). During 2008-2015, 318 ABO-CLKTx were performed at the Department of Surgery III at Tokyo Women's Medical University Hospital. To reduce confounding factors, we adopted a propensity score analysis, which was applied with adjustment for age, gender, duration of pretransplant dialysis, HLA mismatch count, preformed DSA, non-insulin-dependent diabetes mellitus, immunosuppressive treatment, and estimated glomerular filtration rate (eGFR) on postoperative day 7. Using a propensity score matching model (1:1, 115 pairs), we analyzed the long-term outcomes of 230 ABO-CLKTx recipients retrospectively. Recipients were classified into a rituximab-treated (RTX-KTx, N = 115) group and a control group not treated with rituximab (C-KTx, N = 115). During five years, adverse events, survival rates for grafts and patients, and incidence of biopsy-proven acute rejection (BPAR) and dnDSA production for the two groups were monitored and compared. All recipients in the RTX-KTx group received rituximab induction on preoperative day 4 at a single fixed low dose of 100 mg; the CD19+ B cells were eliminated completely before surgery. Of those recipients, 13 (11.3%) developed BPAR; 1 (0.8%) experienced graft loss. By contrast, of C-KTx group recipients, 25 (21.7%) developed BPAR; 3 (2.6%) experienced graft loss. The RTX-KTx group exhibited a significantly lower incidence of BPAR (P = .041) and dnDSA production (13.9% in the RTX-KTx group vs. 26.9% in the C-RTx group, P = .005). Furthermore, lower incidence of CMV infection was detected in the RTX-KTx group than in the C-KTx group (13.9% in the RTX-KTx group vs. 27.0% in the C-KTx group, P = .014). No significant difference was found between groups for several other factors: renal function (P = .384), graft and patient survival (P = .458 and P = .119, respectively), and the respective incidences of BK virus infection (P = .722) and leukopenia (P = .207). During five-year follow-up, single fixed low-dose rituximab therapy is sufficient for ensuring safety, reducing rejection, and suppressing dnDSA production for immunological low-risk non-sensitized ABO-CLKTx.
Collapse
Affiliation(s)
- Yusuke Tomita
- Department of Surgery, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
- * E-mail:
| | - Kazuhiro Iwadoh
- Department of Surgery, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Yuichi Ogawa
- Department of Surgery, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Katsuyuki Miki
- Department of Surgery, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Yojiro Kato
- Department of Surgery, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Kotaro Kai
- Department of Surgery, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Akihito Sannomiya
- Department of Surgery, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Ichiro Koyama
- Department of Surgery, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Kumiko Kitajima
- Department of Surgery, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Ichiro Nakajima
- Department of Surgery, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Shohei Fuchinoue
- Department of Surgery, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| |
Collapse
|
5
|
Tillmann FP, Quack I, Woznowski M, Rump LC. Effect of recipient-donor sex and weight mismatch on graft survival after deceased donor renal transplantation. PLoS One 2019; 14:e0214048. [PMID: 30925158 PMCID: PMC6440625 DOI: 10.1371/journal.pone.0214048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 03/06/2019] [Indexed: 12/05/2022] Open
Abstract
This study evaluated the combined effect of recipient-to-donor weight and sex mismatch after deceased-donor renal transplantation in a German transplant cohort and the evolution of recipient-to-donor weight difference over a 13-year observation period. The association of absolute weight and sex difference with graft failure was explored in an outpatient cohort of deceased-donor transplant recipients who underwent kidney transplantation between 2000 and 2012. Graft failure was defined as repeated need for dialysis or death with a functioning graft. Recipient and donor sex pairings were classified as sex concordant (MDMR/FDFR) or discordant (MDFR/FDMR). These classes were further stratified into four groups according to recipient-to-donor weight mismatch ≥10 kg (recipient > donor) or <10 kg (recipient < donor). Multivariable Cox proportional hazards models were applied to evaluate the time to graft loss adjusting for donor, immunologic, surgical, organizational, and recipient predictors. Sex-concordant transplant pairings <10 kg weight difference served as the reference group. Among 826 transplant recipients, 154 developed graft failure (18.6%). Median graft survival time was 3.9 years; first quartile (0.2-1.2), second quartile (1.2-2.9), third quartile (2.9-5.8), and fourth quartile (5.8-12.4). After multivariable adjustment, the highest relative hazard for graft failure was observed for sex-discordant transplant pairings with a ≥10 kg weight difference between recipient and donor (compared to the reference group MDMR/FDFR with weight difference <10 kg, MDMR/FDFR with weight difference ≥10 kg, hazard ratio 1.86, 95% confidence interval 1.07-3.32-p = 0.029; MDFR/FDMR with weight difference <10 kg, hazard ratio 1.14, 95% confidence interval 0.78-1.68-p = 0.507, and MDFR/FDMR with weight difference ≥10 kg, hazard ratio 2.00, 95% confidence interval 1.15-3.48-p = 0.014). A recipient-to-donor weight mismatch of ≥10 kg was associated with an increased risk of graft loss or recipient death with a functioning graft. Concurrent sex discordance seemed to enhance this effect as indicated by an increase in the hazard ratio. We detected no significant tendency for increasing recipient-to-donor weight differences from 2000 to 2012.
Collapse
Affiliation(s)
- Frank-Peter Tillmann
- Klinik für Nephrologie, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany
| | - Ivo Quack
- Klinik für Nephrologie, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany
| | - Magdalena Woznowski
- Klinik für Nephrologie, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany
| | - Lars Christian Rump
- Klinik für Nephrologie, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
6
|
Zhao B, Han H, Zhen J, Yang X, Shang J, Xu L, Wang R. CD80 and CTLA-4 as diagnostic and prognostic markers in adult-onset minimal change disease: a retrospective study. PeerJ 2018; 6:e5400. [PMID: 30083478 PMCID: PMC6078067 DOI: 10.7717/peerj.5400] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/18/2018] [Indexed: 12/11/2022] Open
Abstract
Background Minimal change disease (MCD) is a form of idiopathic nephrotic syndrome. Compared to children, adult-onset MCD patients are reported to have delayed responses to glucocorticoid treatment. Several studies of children have suggested detecting urinary CD80 levels to diagnose MCD. There are no effective diagnostic methods to distinguish steroid-sensitive MCD from steroid-resistant MCD unless treatments are used. Methods In a total of 55 patients with biopsy-proven MCD and 26 patients with biopsy-proven idiopathic membranous nephropathy, CD80 and cytotoxic T-lymphocyte antigen-4 (CTLA-4) levels in serum, urine and renal tissue were analyzed. Results Steroid-sensitive MCD patients in remission had lower urinary CD80 levels and higher CTLA-4 levels than patients in relapse (156.65 ± 24.62 vs 1066.40 ± 176.76 ng/g creatinine; p < 0.0001), (728.73 ± 89.93 vs 151.70 ± 27.01 ng/g creatinine; p < 0.0001). For MCD patients in relapse, mean urinary CD80 level was higher, and CTLA-4 level was lower for those who were steroid-sensitive than those who were steroid-resistant (1066.40 ± 176.76 vs. 203.78 ± 30.65 ng/g creatinine; p < 0.0001), but the mean urinary CTLA-4 level was lower (151.70 ± 27.01 vs. 457.83 ± 99.45 ng/g creatinine; p < 0.0001). CD80 expression in glomeruli was a sensitive marker to diagnose MCD. The absent or minimal expression of CTLA-4 in glomeruli could distinguish steroid-sensitive MCD from steroid-resistant MCD. Conclusions Glucocorticoid treatment may result in complete remission for only MCD patients with strongly positive CD80 expression and negative CTLA-4 expression in glomeruli, or higher urinary CD80 level and lower CTLA-4 level.
Collapse
Affiliation(s)
- Bing Zhao
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Hui Han
- Department of Intensive Care Unit, Shandong University Qilu Hospital, Jinan, China
| | - Junhui Zhen
- School of Medicine, Shandong University, Jinan, China
| | - Xiaowei Yang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Jin Shang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liang Xu
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Rong Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
7
|
Burkhalter F, Schaub S, Bucher C, Gürke L, Bachmann A, Hopfer H, Dickenmann M, Steiger J, Binet I. A Comparison of Two Types of Rabbit Antithymocyte Globulin Induction Therapy in Immunological High-Risk Kidney Recipients: A Prospective Randomized Control Study. PLoS One 2016; 11:e0165233. [PMID: 27855166 PMCID: PMC5113896 DOI: 10.1371/journal.pone.0165233] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 10/04/2016] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Induction treatment with rabbit polyclonal antithymocyte globulins (ATGs) is frequent used in kidney transplant recipients with donorspecific HLA antibodies and shows acceptable outcomes. The two commonly used ATGs, Thymoglobulin and ATG-F have slightly different antigen profile and antibody concentrations. The two compounds have never been directly compared in a prospective trial in immunological high-risk recipients. Therefore we performed a prospective randomized controlled study comparing the two compounds in immunological high-risk kidney recipients in terms of safety and efficacy. METHODS Immunological high-risk kidney recipients, defined as the presence of HLA DSA but negative CDC-B and T-cell crossmatches were randomized 1:1 to receive ATG-F or Thymoglobulin. Maintenance immunosuppressive therapy consisted of tacrolimus, mycophenolate mofetil and steroids. RESULTS The per-protocol analysis included 35 patients. There was no immediate infusion reaction observed with both compounds. No PTLD or malignancy occurred during the follow-up in both groups. The incidence of viral and bacterial infections was similar in both groups (p = 0.62). The cumulative incidence of clinical and subclinical antibody mediated allograft rejection as well as T-cell mediated allograft rejection during the first year between ATG-F and Thymoglobulin was similar (35% versus 19%; p = 0.30 and 11% versus 18%; 0.54 respectively). The two-year graft function was similar with a median eGFR of 56 ml/min/1.73m2 (range 21-128) (ATG-F-group) and 51 ml/min/1.73m2 (range 22-132) (Thymo-group) (p = 0.69). CONCLUSION We found no significant differences between the compared study drugs for induction treatment in immunological high-risk patients regarding safety and efficacy during follow-up with good allograft function at 2 years after transplantation.
Collapse
Affiliation(s)
- F Burkhalter
- Clinic for Transplant Immunology and Nephrology, University Hospital Basel, Basel, Switzerland
| | - S Schaub
- Clinic for Transplant Immunology and Nephrology, University Hospital Basel, Basel, Switzerland
| | - Ch Bucher
- Nephrology and Transplantation Medicine, Kantonsspital St Gallen, St Gallen, Switzerland
| | - L Gürke
- Department of Vascular and Transplant Surgery, University Hospital Basel, Basel, Switzerland
| | - A Bachmann
- Department of Urology, Basel University Hospital, Basel, Switzerland
| | - H Hopfer
- Institute for Pathology, University Hospital Basel, Basel, Switzerland
| | - M Dickenmann
- Clinic for Transplant Immunology and Nephrology, University Hospital Basel, Basel, Switzerland
| | - J Steiger
- Clinic for Transplant Immunology and Nephrology, University Hospital Basel, Basel, Switzerland
| | - I Binet
- Nephrology and Transplantation Medicine, Kantonsspital St Gallen, St Gallen, Switzerland
| |
Collapse
|
8
|
Herr F, Brunel M, Roders N, Durrbach A. Co-stimulation Blockade Plus T-Cell Depletion in Transplant Patients: Towards a Steroid- and Calcineurin Inhibitor-Free Future? Drugs 2016; 76:1589-1600. [DOI: 10.1007/s40265-016-0656-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
9
|
Diehl R, Ferrara F, Müller C, Dreyer AY, McLeod DD, Fricke S, Boltze J. Immunosuppression for in vivo research: state-of-the-art protocols and experimental approaches. Cell Mol Immunol 2016; 14:146-179. [PMID: 27721455 PMCID: PMC5301156 DOI: 10.1038/cmi.2016.39] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 05/30/2016] [Accepted: 05/30/2016] [Indexed: 02/06/2023] Open
Abstract
Almost every experimental treatment strategy using non-autologous cell, tissue or organ transplantation is tested in small and large animal models before clinical translation. Because these strategies require immunosuppression in most cases, immunosuppressive protocols are a key element in transplantation experiments. However, standard immunosuppressive protocols are often applied without detailed knowledge regarding their efficacy within the particular experimental setting and in the chosen model species. Optimization of such protocols is pertinent to the translation of experimental results to human patients and thus warrants further investigation. This review summarizes current knowledge regarding immunosuppressive drug classes as well as their dosages and application regimens with consideration of species-specific drug metabolization and side effects. It also summarizes contemporary knowledge of novel immunomodulatory strategies, such as the use of mesenchymal stem cells or antibodies. Thus, this review is intended to serve as a state-of-the-art compendium for researchers to refine applied experimental immunosuppression and immunomodulation strategies to enhance the predictive value of preclinical transplantation studies.
Collapse
Affiliation(s)
- Rita Diehl
- Fraunhofer-Institute for Cell Therapy and Immunology, Leipzig 04103, Germany
| | - Fabienne Ferrara
- Fraunhofer-Institute for Cell Therapy and Immunology, Leipzig 04103, Germany.,Institute of Vegetative Physiology, Charite University Medicine and Center for Cardiovascular Research, Berlin 10115, Germany
| | - Claudia Müller
- Fraunhofer-Institute for Cell Therapy and Immunology, Leipzig 04103, Germany
| | - Antje Y Dreyer
- Fraunhofer-Institute for Cell Therapy and Immunology, Leipzig 04103, Germany
| | | | - Stephan Fricke
- Fraunhofer-Institute for Cell Therapy and Immunology, Leipzig 04103, Germany
| | - Johannes Boltze
- Fraunhofer-Institute for Cell Therapy and Immunology, Leipzig 04103, Germany.,Fraunhofer Research Institution for Marine Biotechnology and Institute for Medical and Marine Biotechnology, University of Lübeck, Lübeck 23562, Germany
| |
Collapse
|
10
|
Bolton HA, Zhu E, Terry AM, Guy TV, Koh WP, Tan SY, Power CA, Bertolino P, Lahl K, Sparwasser T, Shklovskaya E, Fazekas de St Groth B. Selective Treg reconstitution during lymphopenia normalizes DC costimulation and prevents graft-versus-host disease. J Clin Invest 2015; 125:3627-41. [PMID: 26301814 DOI: 10.1172/jci76031] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 07/13/2015] [Indexed: 01/03/2023] Open
Abstract
Regulatory T cells (Tregs) have been shown to enhance immune reconstitution and prevent graft-versus-host disease (GVHD) after hematopoietic stem cell transplantation; however, it is unclear how Tregs mediate these effects. Here, we developed a model to examine the mechanism of Treg-dependent regulation of immune reconstitution. Lymphopenic mice were selectively reconstituted with Tregs prior to transfer of conventional CD4+ T cells. Full Treg reconstitution prevented the rapid oligoclonal proliferation that gives rise to pathogenic CD4 effector T cells, while preserving the slow homeostatic form of lymphopenia-induced peripheral expansion that repopulates a diverse peripheral T cell pool. Treg-mediated CTLA-4-dependent downregulation of CD80/CD86 on DCs was critical for inhibition of rapid proliferation and was a function of the Treg/DC ratio achieved by reconstitution. In an allogeneic BM transplant model, selective Treg reconstitution before T cell transfer also normalized DC costimulation and provided complete protection against GVHD. In contrast, cotransfer of Tregs was not protective. Our results indicate that achieving optimal recovery from lymphopenia should aim to improve early Treg reconstitution in order to increase the relative number of Tregs to DCs and thereby inhibit spontaneous oligoclonal T cell proliferation.
Collapse
|
11
|
You S. Differential sensitivity of regulatory and effector T cells to cell death: a prerequisite for transplant tolerance. Front Immunol 2015; 6:242. [PMID: 26042125 PMCID: PMC4437185 DOI: 10.3389/fimmu.2015.00242] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 05/06/2015] [Indexed: 12/13/2022] Open
Abstract
Despite significant progress achieved in transplantation, immunosuppressive therapies currently used to prevent graft rejection are still endowed with severe side effects impairing their efficiency over the long term. Thus, the development of graft-specific, non-toxic innovative therapeutic strategies has become a major challenge, the goal being to selectively target alloreactive effector T cells while sparing CD4+Foxp3+ regulatory T cells (Tregs) to promote operational tolerance. Various approaches, notably the one based on monoclonal antibodies or fusion proteins directed against the TCR/CD3 complex, TCR coreceptors, or costimulatory molecules, have been proposed to reduce the alloreactive T cell pool, which is an essential prerequisite to create a therapeutic window allowing Tregs to induce and maintain allograft tolerance. In this mini review, we focus on the differential sensitivity of Tregs and effector T cells to the depleting and inhibitory effect of these immunotherapies, with a particular emphasis on CD3-specific antibodies that beyond their immunosuppressive effect, also express potent tolerogenic capacities.
Collapse
Affiliation(s)
- Sylvaine You
- Université Paris Descartes, Sorbonne Paris Cité , Paris , France ; INSERM U1151, Institut Necker-Enfants Malades , Paris , France ; CNRS UMR 8253, Institut Necker-Enfants Malades , Paris , France
| |
Collapse
|
12
|
Soluble co-signaling molecules predict long-term graft outcome in kidney-transplanted patients. PLoS One 2014; 9:e113396. [PMID: 25478957 PMCID: PMC4257538 DOI: 10.1371/journal.pone.0113396] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 10/23/2014] [Indexed: 12/25/2022] Open
Abstract
Co-signaling molecules are responsible for full T-cell activation after solid organ transplantation. Their increased expression can lead to the release of a soluble form that can modulate the immune response post-transplantation. We analyzed the presence of co-signaling molecules (sCD30, sCD40, sCD137, sCTLA-4, sCD80, sCD28, sCD40L, sPD-1, and sPD-L1) in serum from kidney-transplanted patients (n = 59) obtained at different times (before transplantation, and 15 days, 3 months and 1 year post-transplantation) and their contribution to graft outcome was evaluated using principal component analysis. Before transplantation, high levels of soluble co-signaling molecules (mainly sCD30, sCD137 and sCD40) were detected in all patients. These molecules were modulated soon after receiving an allograft but never attained similar levels to those of healthy controls. A signature based on the determination of six soluble co-stimulatory (sCD30, sCD40, sCD137 and sCD40L) and co-inhibitory (sPD-1 and sPD-L1) molecules at 3 months post-transplantation allowed a group of patients to be identified (27.12%) with a worse long-term graft outcome. Patients with high levels of soluble molecules showed a progressive and gradual deterioration of kidney function (increased creatinine and proteinuria levels and decreased estimated glomerular filtration rate) over time and a higher risk of graft loss at 6 years post-transplantation than patients with low levels of these molecules (62.55% versus 5.14%, p<0.001). Thus, our data show an aberrant expression of soluble co-signaling molecules in kidney-transplanted patients whose quantification at 3 months post-transplantation might be a useful biomarker of immune status and help to predict long-term graft evolution.
Collapse
|
13
|
Immunophenotyping and efficacy of low dose ATG in non-sensitized kidney recipients undergoing early steroid withdrawal: a randomized pilot study. PLoS One 2014; 9:e104408. [PMID: 25111080 PMCID: PMC4128673 DOI: 10.1371/journal.pone.0104408] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 07/03/2014] [Indexed: 01/30/2023] Open
Abstract
Rabbit antithymocyte globulin (ATG) is commonly used as an induction therapy in renal transplant recipients, but the ideal dosage in tacrolimus-based early steroid withdrawal protocols has not been established. The purpose of this pilot study was to determine the immunophenotyping and efficacy of lower dose ATG in low immunological-risk kidney transplant recipients. In this prospective study, 45 patients were randomized (1∶1) to our standard dose ATG (total dose 3.75 mg/kg)(sATG) vs. lower dose 2.25 mg/kg (lowATG). All patients underwent early steroid withdrawal within 7 days. The primary end point was biopsy-proven acute rejection at 12 months. Prospective immunophenotyping of freshly isolated PBMCs was performed at baseline, 3, 6, 12 months post-transplant. The rate of acute rejection was 17% and 10% in the sATG and lowATG, respectively. Effector memory T cells, Tregs and recent thymic emigrants T cells had similar kinetics post-transplant in both groups. No statistically significant differences were found in graft survival, patient survival or infections between the two groups, though there was a non-significant increase in leukopenia (43%v s. 30%), CMV (8% vs. 0) and BK (4% vs. 0) infections in sATG group vs. lowATG. In sum, in low immunological risk kidney recipients undergoing steroid withdrawal, low dose ATG seems to be efficacious in preventing acute rejection and depleting T cells with potentially lower infectious complications. A larger study is warranted to confirm these findings. Trial Registration ClinicalTrials.gov NCT00548405
Collapse
|