1
|
Yue L, Li J, Yao M, Song S, Zhang X, Wang Y. Cutting edge of immune response and immunosuppressants in allogeneic and xenogeneic islet transplantation. Front Immunol 2024; 15:1455691. [PMID: 39346923 PMCID: PMC11427288 DOI: 10.3389/fimmu.2024.1455691] [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: 06/27/2024] [Accepted: 08/27/2024] [Indexed: 10/01/2024] Open
Abstract
As an effective treatment for diabetes, islet transplantation has garnered significant attention and research in recent years. However, immune rejection and the toxicity of immunosuppressive drugs remain critical factors influencing the success of islet transplantation. While immunosuppressants are essential in reducing immune rejection reactions and can significantly improve the survival rate of islet transplants, improper use of these drugs can markedly increase mortality rates following transplantation. Additionally, the current availability of islet organ donations fails to meet the demand for organ transplants, making xenotransplantation a crucial method for addressing organ shortages. This review will cover the following three aspects: 1) the immune responses occurring during allogeneic islet transplantation, including three stages: inflammation and IBMIR, allogeneic immune response, and autoimmune recurrence; 2) commonly used immunosuppressants in allogeneic islet transplantation, including calcineurin inhibitors (Cyclosporine A, Tacrolimus), mycophenolate mofetil, glucocorticoids, and Bortezomib; and 3) early and late immune responses in xenogeneic islet transplantation and the immune effects of triple therapy (ECDI-fixed donor spleen cells (ECDI-SP) + anti-CD20 + Sirolimus) on xenotransplantation.
Collapse
Affiliation(s)
- Liting Yue
- Center of Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jisong Li
- Department of Gastrointestinal Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Mingjun Yao
- Center of Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Xiaoqin Zhang
- Center of Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yi Wang
- Center of Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, Chengdu, China
| |
Collapse
|
2
|
Abstract
Acute graft-versus-host disease (GVHD), the major complication after allogeneic hematopoietic cell transplant (HCT), develops in approximately 50% of patients. The primary treatment is high-dose systemic steroids, but treatment failure is common, and steroid-refractory (SR) GVHD is the leading cause of non-relapse mortality after allogeneic HCT. Ruxolitinib became the first treatment for SR GVHD to obtain US Food and Drug Administration approval, and other new treatments are actively being studied. We searched the literature using the PubMed database and clinical trials using ClinicalTrials.gov to identify the most promising new treatments for GVHD. In this review, we categorize potential new treatments for GVHD by their mechanism of action (e.g., antibodies that deplete T cells or prevent their trafficking to target tissues, proteasome inhibitors, tyrosine kinase inhibitors, and other agents) and summarize the results from clinical trials.
Collapse
Affiliation(s)
- Stelios Kasikis
- Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA
| | - Aaron Etra
- Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA
| | - John E Levine
- Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA.
| |
Collapse
|
3
|
A combination regimen of low-dose bortezomib and rapamycin prolonged the graft survival in a murine allogeneic islet transplantation model. Immunol Lett 2019; 216:21-27. [PMID: 31593743 DOI: 10.1016/j.imlet.2019.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/27/2019] [Accepted: 10/02/2019] [Indexed: 11/21/2022]
Abstract
As the first FDA-approved proteasome inhibitor drug, bortezomib has been used for the treatment of multiple myeloma and lymphoma. However, its effects alone or in combination with other immunosuppressants on allogeneic islet transplantation have not been reported so far. In this study, we showed that the short-term combination treatment of low-dose bortezomib and rapamycin significantly prolonged the survival of islet allografts. Short-term treatment of low-dose (0.05 mg/kg or 0.1 mg/kg) bortezomib reduced the MHC class II expression in dendritic cells (DCs) of alloantigen-sensitized mice, and prolonged the islet allograft survival for up to 50 days in diabetic mice. Notably, when bortezomib was combined with rapamycin, it induced islet-specific immunological tolerance which allowed the acceptance of a second graft without additional immunosuppression. This regimen dramatically reduced the alloantigen-specific IFN-γ-producing T cells in the spleen, and increased regulatory T cells both at the graft site and in the spleen. Therefore, we propose that short-term treatment of low-dose bortezomib and rapamycin could be a new tolerance-promoting immunosuppressive regimen for allogeneic islet transplantation.
Collapse
|
4
|
Abstract
Introduction: Graft-versus-host disease (GVHD) is the most common complication of hematopoietic stem cell transplantation (HSCT); therefore, the prevention of GVHD is important for a successful treatment. Tacrolimus (Tac), a calcineurin inhibitor, has been widely used for the prophylaxis of GVHD in HSCT recipients. Areas covered: This review introduces phase II/III of clinical trials related with Tac's roles in the prevention of GVHD in HSCT. Furthermore, we discuss the normal ranges of Tac concentrations, pharmacogenetics, and drug interactions of Tac, as well as its side effects in adult HSCT recipients. Expert opinion: A series of studies has established the efficacy and safety of Tac alone or in combination with other agents in HSCT. However, successful administration of Tac is complicated by its narrow therapeutic window, inter-patient pharmacokinetic variability, and a spectrum of undesirable side effects. It is necessary to maintain concentrations of Tac within the desired ranges for GVHD prophylaxis. Moreover, various factors contribute to significant variability in Tac pharmacokinetics, including drug interactions and genomic variation.
Collapse
Affiliation(s)
- Yuan Gao
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Soochow University , Suzhou , China
| | - Jingjing Ma
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Soochow University , Suzhou , China
| |
Collapse
|
5
|
Phase I study of graft-versus-host disease prophylaxis including bortezomib for allogeneic hematopoietic cell transplantation from unrelated donors with one or two HLA loci mismatches in Japanese patients. Int J Hematol 2019; 110:736-742. [PMID: 31560116 DOI: 10.1007/s12185-019-02743-6] [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: 07/12/2019] [Revised: 09/19/2019] [Accepted: 09/19/2019] [Indexed: 10/25/2022]
Abstract
This phase I study was designed for graft-versus-host disease (GVHD) prophylaxis including bortezomib in allogeneic hematopoietic cell transplantation (allo-HCT) from human leukocyte antigen (HLA)-mismatched unrelated donors in Japanese patients. Patients were administered bortezomib on days 1, 4, and 7, with short-term methotrexate and tacrolimus. Three bortezomib dose levels were prepared (1.0, 1.3, and 1.5 mg/m2). A dose of 1.3 mg/m2 was planned for administration to the initial six patients, and was adjusted if dose-limiting toxicity developed. Five of six patients enrolled for the initial dose had bone marrow donors. Two cases had single-antigen and single-allele mismatches; four had single-antigen mismatch at the A, B, C, and/or DRB1 loci in the GVH direction. All patients achieved neutrophil engraftment and complete donor chimerism. Three patients developed grade II acute GVHD, and none developed grade III-IV GVHD or any dose-limiting toxicity attributable to bortezomib by day 100. Two patients developed late-onset acute GVHD, and two developed chronic GVHD, but all cases were manageable. All patients were alive without relapse after a median follow-up period of 52 months. The optimal dose of bortezomib was determined to be 1.3 mg/m2. Prophylaxis against GVHD using a regimen including bortezomib thus seems feasible for HLA-mismatched unrelated allo-HCT.
Collapse
|
6
|
MacDonald KPA, Betts BC, Couriel D. Reprint of: Emerging Therapeutics for the Control of Chronic Graft-versus-Host Disease. Biol Blood Marrow Transplant 2018; 24:S7-S14. [PMID: 29425517 DOI: 10.1016/j.bbmt.2017.12.788] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 10/03/2017] [Indexed: 02/02/2023]
Affiliation(s)
- Kelli P A MacDonald
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.
| | - Brian C Betts
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffit Cancer Center, Tampa, Florida
| | - Daniel Couriel
- Department of Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| |
Collapse
|
7
|
Koreth J, Kim HT, Lange PB, Poryanda SJ, Reynolds CG, Rai SC, Armand P, Cutler CS, Ho VT, Glotzbecker B, Yusuf R, Nikiforow S, Chen YB, Dey B, McMasters M, Ritz J, Blazar BR, Soiffer RJ, Antin JH, Alyea EP. Bortezomib-based immunosuppression after reduced-intensity conditioning hematopoietic stem cell transplantation: randomized phase II results. Haematologica 2018; 103:522-530. [PMID: 29326124 PMCID: PMC5830392 DOI: 10.3324/haematol.2017.176859] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 01/10/2018] [Indexed: 11/16/2022] Open
Abstract
Aprior phase I/II trial of bortezomib/tacrolimus/methotrexate prophylaxis after human leukocyte antigen (HLA)-mismatched reduced intensity conditioning allogeneic hematopoietic stem cell transplantation documented low acute graft-versus-host disease incidence, with promising overall and progression-free survival. We performed an open-label three-arm 1:1:1 phase II randomized controlled trial comparing grade II–IV acute graft-versus-host disease between conventional tacrolimus/methotrexate (A) versus bortezomib/tacrolimus/methotrexate (B), and versus bortezomib/sirolimus/tacrolimus (C), in reduced intensity conditioning allogeneic transplantation recipients lacking HLA-matched related donors. The primary endpoint was grade II–IV acute graft-versus-host disease incidence rate by day +180. One hundred and thirty-eight patients (A 46, B 45, C 47) with a median age of 64 years (range: 24–75), varying malignant diagnoses and disease risk (low 14, intermediate 96, high/very high 28) received 7–8/8 HLA-mismatched (40) or matched unrelated donor (98) grafts. Median follow up in survivors was 30 months (range: 14–46). Despite early immune reconstitution differences, day +180 grade II-IV acute graft-versus-host disease rates were similar (A 32.6%, B 31.1%, C 21%; P=0.53 for A vs. B, P=0.16 for A vs. C). The 2-year non-relapse mortality incidence was similar (A 14%, B 16%, C 6.4%; P=0.62), as were relapse (A 32%, B 32%, C 38%; P=0.74), chronic graft-versus-host disease (A 59%, B 60% C 55%; P=0.66), progression-free survival (A 54%, B 52%, C 55%; P=0.95), and overall survival (A 61%, B 62%, C 62%; P=0.98). Overall, the bortezomib-based regimens evaluated did not improve outcomes compared with tacrolimus/methotrexate therapy. clinicaltrials.gov Identifier: 01754389
Collapse
Affiliation(s)
- John Koreth
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Haesook T Kim
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard School of Public Health, Boston, MA
| | - Paulina B Lange
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Samuel J Poryanda
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Carol G Reynolds
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Sharmila Chamling Rai
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Philippe Armand
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Corey S Cutler
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Vincent T Ho
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Brett Glotzbecker
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Rushdia Yusuf
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Sarah Nikiforow
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Yi-Bin Chen
- Massachusetts General Hospital Cancer Center, Boston, MA
| | | | | | - Jerome Ritz
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Bruce R Blazar
- University of Minnesota Masonic Cancer Center and Department of Pediatrics, Division of Blood and Marrow Transplantation, Minneapolis, MN, USA
| | - Robert J Soiffer
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Joseph H Antin
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Edwin P Alyea
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| |
Collapse
|
8
|
Emerging Therapeutics for the Control of Chronic Graft-versus-Host Disease. Biol Blood Marrow Transplant 2017; 24:19-26. [PMID: 29032060 DOI: 10.1016/j.bbmt.2017.10.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 10/03/2017] [Indexed: 02/01/2023]
|
9
|
Im A, Hakim FT, Pavletic SZ. Novel targets in the treatment of chronic graft-versus-host disease. Leukemia 2016; 31:543-554. [PMID: 27899803 DOI: 10.1038/leu.2016.367] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 11/14/2016] [Accepted: 11/16/2016] [Indexed: 12/19/2022]
Abstract
Despite advances that have improved survival after allogeneic hematopoietic stem cell transplantation (HCT), chronic graft-versus-host disease (GVHD) remains a leading cause of late morbidity and mortality after transplant. Current treatment options show limited efficacy in steroid-refractory disease, and there exists a paucity of robust data to guide management decisions. Lack of United States Food and Drug Administration (FDA)- or European Medicines Agency (EMA)-approved agents in GVHD underscore the importance of developing novel therapies. Better understanding of the biology of chronic GVHD has provided novel targets for treatment, and structured guidelines in diagnosis and in clinical trial design have provided a common language and pathways for research in this area. These, combined with the surge of drug development in Oncology and Immunology, are factors that have contributed to the accelerating field of drug development and clinical research in chronic GVHD. In these exciting times, it is possible to foresee long awaited advances in the treatment of this devastating complication of HCT. This review will summarize the ongoing clinical development for novel therapies in chronic GVHD.
Collapse
Affiliation(s)
- A Im
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh Cancer Institute and UPMC Cancer Centers, Pittsburgh, PA, USA.,Experimental Transplantation and Immunology Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD, USA
| | - F T Hakim
- Experimental Transplantation and Immunology Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD, USA
| | - S Z Pavletic
- Experimental Transplantation and Immunology Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
10
|
Mechanistic approaches for the prevention and treatment of chronic GVHD. Blood 2016; 129:22-29. [PMID: 27821505 DOI: 10.1182/blood-2016-08-686659] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/11/2016] [Indexed: 02/07/2023] Open
Abstract
Clinical outcomes for patients undergoing allogeneic hematopoietic stem cell transplantation continue to improve, but chronic graft-versus-host disease (GVHD) remains a common toxicity and major cause of nonrelapse morbidity and mortality. Treatment of chronic GVHD has previously relied primarily on corticosteroids and other broadly immune suppressive agents. However, conventional immune suppressive agents have limited clinical efficacy in chronic GVHD, and prolonged immune suppressive treatments result in additional toxicities that further limit clinical recovery from transplant and return to normal daily function. Recent advances in our understanding of the immune pathology of chronic GVHD offer the possibility that new therapeutic approaches can be directed in more precise ways to target specific immunologic mechanisms and pathways. In this review, we briefly summarize current standard treatment options and present new therapeutic approaches that are supported by preclinical studies and early-phase clinical trials suggesting that these approaches may have clinical utility for treatment or prevention of chronic GVHD. Further evaluation of these new therapeutic options in well-designed prospective multicenter trials are needed to identify the most effective new agents and improve outcomes for patients with chronic GVHD.
Collapse
|
11
|
Ng ZY, Read C, Kurtz JM, Cetrulo CL. Memory T Cells in Vascularized Composite Allotransplantation. ACTA ACUST UNITED AC 2016. [DOI: 10.1080/23723505.2016.1229649] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
12
|
Pearl MH, Nayak AB, Ettenger RB, Puliyanda D, Palma Diaz MF, Zhang Q, Reed EF, Tsai EW. Bortezomib may stabilize pediatric renal transplant recipients with antibody-mediated rejection. Pediatr Nephrol 2016; 31:1341-8. [PMID: 27048228 PMCID: PMC5590841 DOI: 10.1007/s00467-016-3319-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 12/28/2015] [Accepted: 12/30/2015] [Indexed: 01/20/2023]
Abstract
BACKGROUND Current therapeutic strategies to effectively treat antibody-mediated rejection (AMR) are insufficient. Thus, we aimed to determine the benefit of a therapeutic protocol using bortezomib for refractory C4d + AMR in pediatric kidney transplant patients. METHODS We examined seven patients with treatment-refractory C4d + AMR. Immunosuppression included antithymocyte globulin or anti-CD25 monoclonal antibody for induction therapy with maintenance corticosteroids, calcineurin inhibitor, and anti-metabolite. Estimated glomerular filtration rate (eGFR) calculated by the Schwartz equation, biopsy findings assessed by 2013 Banff criteria, and human leukocyte antigen (HLA) donor-specific antibodies (DSA) performed using the Luminex single antigen bead assay were monitored pre- and post- bortezomib therapy. RESULTS Seven patients (86 % male, 86 % with ≥6/8 HLA mismatch, and 14 % with pre-formed DSA) age 5 to 19 (median 15) years developed refractory C4d + AMR between 1 and 145 (median 65) months post-transplantation. All patients tolerated bortezomib. One patient had allograft loss. Of the six patients with surviving grafts (86 %), mean pre-bortezomib eGFR was 42 ml/min/1.73 m(2) and the mean 1 year post-bortezomib eGFR was 53 ml/min/1.73 m(2). Five of seven (71 %) had improvement of histological findings of AMR, C4d staining, and/or acute cellular rejection. Reduction in HLA DSAs was more effective for class I than class II. CONCLUSIONS Bortezomib appears safe and may correlate with stabilization of eGFR in pediatric kidney transplant patients with refractory C4d + AMR.
Collapse
Affiliation(s)
- Meghan H Pearl
- Department of Pediatrics, Division of Nephrology, David Geffen School of Medicine at UCLA, University of California Los Angeles, PO Box 951752, Los Angeles, CA, 90095, USA.
| | - Anjali B Nayak
- Department of Pediatrics, Division of Nephrology, David Geffen School of Medicine at UCLA, University of California Los Angeles, PO Box 951752, Los Angeles, CA, 90095, USA
| | - Robert B Ettenger
- Department of Pediatrics, Division of Nephrology, David Geffen School of Medicine at UCLA, University of California Los Angeles, PO Box 951752, Los Angeles, CA, 90095, USA
| | - Dechu Puliyanda
- Pediatric Nephrology and Transplant Immunology, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Miguel Fernando Palma Diaz
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, PO Box 951752, Los Angeles, CA, USA
| | - Qiuheng Zhang
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, PO Box 951752, Los Angeles, CA, USA
| | - Elaine F Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, PO Box 951752, Los Angeles, CA, USA
| | - Eileen W Tsai
- Department of Pediatrics, Division of Nephrology, David Geffen School of Medicine at UCLA, University of California Los Angeles, PO Box 951752, Los Angeles, CA, 90095, USA
| |
Collapse
|
13
|
Abstract
INTRODUCTION Graft-versus-host disease (GVHD) leads to significant morbidity and mortality after allogeneic stem cell transplantation. While corticosteroids alone are adequate in some cases, they are often insufficient, leading to poor quality of life associated with the symptoms of disease, or mortality from infection and GVHD. Moreover, corticosteroids have significant side effects and often do not lead to durable responses. New therapies are needed to improve the development and progression of acute and chronic GVHD. AREAS COVERED We discuss the spectrum of emerging drugs for GVHD prevention and therapy. Cellular therapies will be briefly discussed. The available pre-clinical and clinical data regarding monoclonal antibodies, interleukin-2, alpha-1 antitrypsin, histone deacetylase inhibitors, tyrosine kinase inhibitors, and proteasome inhibitors will be reviewed. EXPERT OPINION Although therapies emerging for GVHD remain promising, most of these drugs are still in early phase clinical trials and require randomized comparisons before formal conclusions can be drawn. It is likely that in the near future some of these agents will show improvements in response when compared with corticosteroids alone. Although it is difficult to predict which of these agents will be most promising, alpha-1 antitrypsin, ruxolitinib and interleukin-2 have demonstrated encouraging results.
Collapse
Affiliation(s)
- Natasha Kekre
- a Division of Hematology , Ottawa Health Research Institute, The Ottawa Hospital and University of Ottawa , Ottawa , ON , Canada
| | - Joseph H Antin
- b Blood and Marrow Transplantation Program, Division of Hematologic Malignancies , Dana-Farber Cancer Institute, Harvard Medical School , Boston , MA , USA
| |
Collapse
|
14
|
Ex vivo evaluation of the effect of regulatory T cells on the anti-tumor activity of bortezomib in multiple myeloma. Exp Hematol 2016; 44:223-30. [DOI: 10.1016/j.exphem.2015.05.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 05/14/2015] [Accepted: 05/29/2015] [Indexed: 11/21/2022]
|
15
|
Merino D, San Segundo D, Medina JM, Rodrigo E, Asensio E, Irure J, Fernández-Fresnedo G, Arias MA, López-Hoyos M. Different in vitro proliferation and cytokine-production inhibition of memory T-cell subsets after calcineurin and mammalian target of rapamycin inhibitors treatment. Immunology 2016; 148:206-15. [PMID: 26931075 DOI: 10.1111/imm.12603] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 02/10/2016] [Accepted: 02/24/2016] [Indexed: 12/14/2022] Open
Abstract
Calcineurin inhibitors (CNI) and mammalian target of rapamycin inhibitors (mTORi) are the main immunosuppressants used for long-term maintenance therapy in transplant recipients to avoid acute rejection episodes. Both groups of immunosuppressants have wide effects and are focused against the T cells, although different impacts on specific T-cell subsets, such as regulatory T cells, have been demonstrated. A greater knowledge of the impact of immunosuppression on the cellular components involved in allograft rejection could facilitate decisions for individualized immunosuppression when an acute rejection event is suspected. Memory T cells have recently gained focus because they might induce a more potent response compared with naive cells. The impact of immunosuppressants on different memory T-cell subsets remains unclear. In the present study, we have studied the specific impact of CNI (tacrolimus) and mTORi (rapamycin and everolimus) over memory and naive CD4(+) T cells. To do so, we have analysed the proliferation, phenotypic changes and cytokine synthesis in vitro in the presence of these immunosuppressants. The present work shows a more potent effect of CNI on proliferation and cytokine production in naive and memory T cells. However, the mTORi permit the differentiation of naive T cells to the memory phenotype and allow the production of interleukin-2. Taken together, our data show evidence to support the combined use of CNI and mTORi in transplant immunosuppression.
Collapse
Affiliation(s)
- David Merino
- Autoimmunity and Transplant Group-IDIVAL, Santander, Spain
| | - David San Segundo
- Immunology Service Hospital Universitario Marqués de Valdecilla-IDIVAL, Santander, Spain
| | - Juan M Medina
- Autoimmunity and Transplant Group-IDIVAL, Santander, Spain
| | - Emilio Rodrigo
- Nephrology Service Hospital Universitario Marqués de Valdecilla-IDIVAL, Santander, Spain
| | - Esther Asensio
- Immunology Service Hospital Universitario Marqués de Valdecilla-IDIVAL, Santander, Spain
| | - Juan Irure
- Immunology Service Hospital Universitario Marqués de Valdecilla-IDIVAL, Santander, Spain
| | | | - Manuel A Arias
- Nephrology Service Hospital Universitario Marqués de Valdecilla-IDIVAL, Santander, Spain
| | - Marcos López-Hoyos
- Immunology Service Hospital Universitario Marqués de Valdecilla-IDIVAL, Santander, Spain
| |
Collapse
|
16
|
Lin K, Chen S, Chen G. Role of Memory T Cells and Perspectives for Intervention in Organ Transplantation. Front Immunol 2015; 6:473. [PMID: 26441978 PMCID: PMC4568416 DOI: 10.3389/fimmu.2015.00473] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 08/31/2015] [Indexed: 12/12/2022] Open
Abstract
Memory T cells are necessary for protective immunity against invading pathogens, especially under conditions of immunosuppression. However, their presence also threatens transplant survival, making transplantation a great challenge. Significant progress has been achieved in recent years in advancing our understanding of the role that memory T cells play in transplantation. This review focuses on the latest advances in our understanding of the involvement of memory T cells in graft rejection and transplant tolerance and discusses potential strategies for targeting memory T cells in order to minimize allograft rejection and optimize clinical outcomes.
Collapse
Affiliation(s)
- Kailin Lin
- Institute of Organ Transplantation, Tongji Hospital, Huazhong University of Science and Technology , Wuhan , China
| | - Song Chen
- Institute of Organ Transplantation, Tongji Hospital, Huazhong University of Science and Technology , Wuhan , China ; Key Laboratory of Organ Transplantation, Ministry of Education , Wuhan , China ; Key Laboratory of Organ Transplantation, Ministry of Public Health , Wuhan , China
| | - Gang Chen
- Institute of Organ Transplantation, Tongji Hospital, Huazhong University of Science and Technology , Wuhan , China ; Key Laboratory of Organ Transplantation, Ministry of Education , Wuhan , China ; Key Laboratory of Organ Transplantation, Ministry of Public Health , Wuhan , China
| |
Collapse
|
17
|
Koreth J, Kim HT, Lange PB, Bindra B, Reynolds CG, Chammas MJ, Armand P, Cutler CS, Ho VT, Glotzbecker B, Nikiforow S, Ritz J, Blazar BR, Soiffer RJ, Antin JH, Alyea EP. A Bortezomib-Based Regimen Offers Promising Survival and Graft-versus-Host Disease Prophylaxis in Myeloablative HLA-Mismatched and Unrelated Donor Transplantation: A Phase II Trial. Biol Blood Marrow Transplant 2015; 21:1907-13. [PMID: 26055298 DOI: 10.1016/j.bbmt.2015.05.027] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 05/30/2015] [Indexed: 11/25/2022]
Abstract
Hematopoietic stem cell transplantation (HSCT) recipients lacking HLA-matched related donors have increased graft-versus-host disease (GVHD) and nonrelapse mortality (NRM). Bortezomib added to reduced-intensity conditioning can offer benefit in T cell-replete HLA-mismatched HSCT and may also benefit myeloablative conditioning (MAC) transplants. We conducted a phase II trial of short-course bortezomib plus standard tacrolimus/methotrexate after busulfan/fludarabine MAC in 34 patients with predominantly myeloid malignancies. Fourteen (41%) received 8/8 HLA-matched unrelated donor (MUD) and 20 (59%) received 7/8 HLA-mismatched related/unrelated donor peripheral blood stem cell grafts. Median age was 49 years (range, 21 to 60), and median follow-up was 25 months (range, 11 to 36). The regimen was well tolerated. No dose modifications were required. Neutrophil and platelet engraftment occurred at a median of 14 (range, 10 to 33) and 17 (range, 10 to 54) days, respectively. Median 30-day donor chimerism was 99% (range, 90 to 100), and 100-day grades II to IV and III to IV acute GVHD incidence was 32% and 12% respectively. One-year chronic GVHD incidence was 50%. Two-year cumulative incidence of both NRM and relapse was 16%. Two-year progression-free and overall survival rates were 70% and 71%, respectively. Outcomes were comparable to an 8/8 MUD MAC cohort (n = 45). Immune reconstitution was robust. Bortezomib-based MAC HSCT is well tolerated, with HLA-mismatched outcomes comparable with 8/8 MUD MAC HSCT, and is suitable for randomized evaluation. (clinicaltrials.gov: NCT01323920.).
Collapse
Affiliation(s)
- John Koreth
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
| | - Haesook T Kim
- Department of Biostatistics & Computational Biology, Dana-Farber Cancer Institute and Harvard School of Public Health, Boston, Massachusetts
| | - Paulina B Lange
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Bhavjot Bindra
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Carol G Reynolds
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Marie J Chammas
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Philippe Armand
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Corey S Cutler
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Vincent T Ho
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Brett Glotzbecker
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Sarah Nikiforow
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Jerome Ritz
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, University of Minnesota Masonic Cancer Center and Department of Pediatrics, Minneapolis, Minnesota
| | - Robert J Soiffer
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Joseph H Antin
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Edwin P Alyea
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
18
|
Michel SG, Madariaga MLL, Villani V, Shanmugarajah K. Current progress in xenotransplantation and organ bioengineering. Int J Surg 2014; 13:239-244. [PMID: 25496853 DOI: 10.1016/j.ijsu.2014.12.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 11/30/2014] [Accepted: 12/07/2014] [Indexed: 12/25/2022]
Abstract
Organ transplantation represents a unique method of treatment to cure people with end-stage organ failure. Since the first successful organ transplant in 1954, the field of transplantation has made great strides forward. However, despite the ability to transform and save lives, transplant surgery is still faced with a fundamental problem the number of people requiring organ transplants is simply higher than the number of organs available. To put this in stark perspective, because of this critical organ shortage 18 people every day in the United States alone die on a transplant waiting list (U.S. Department of Health & Human Services, http://organdonor.gov/about/data.html). To address this problem, attempts have been made to increase the organ supply through xenotransplantation and more recently, bioengineering. Here we trace the development of both fields, discuss their current status and highlight limitations going forward. Ultimately, lessons learned in each field may prove widely applicable and lead to the successful development of xenografts, bioengineered constructs, and bioengineered xeno-organs, thereby increasing the supply of organs for transplantation.
Collapse
Affiliation(s)
- Sebastian G Michel
- Transplantation Biology Research Center, Massachusetts General Hospital, Building 149, 13th Street, Charlestown, Boston, MA 02114, USA; Department of Cardiac Surgery, Ludwig-Maximilians-Universität München, Munich D-81377, Germany.
| | - Maria Lucia L Madariaga
- Transplantation Biology Research Center, Massachusetts General Hospital, Building 149, 13th Street, Charlestown, Boston, MA 02114, USA; Department of Surgery, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02129, USA
| | - Vincenzo Villani
- Transplantation Biology Research Center, Massachusetts General Hospital, Building 149, 13th Street, Charlestown, Boston, MA 02114, USA
| | - Kumaran Shanmugarajah
- Transplantation Biology Research Center, Massachusetts General Hospital, Building 149, 13th Street, Charlestown, Boston, MA 02114, USA; Division of Surgery, Imperial College London, Exhibition Road, London SW7 2AZ, United Kingdom.
| |
Collapse
|
19
|
May LJ, Yeh J, Maeda K, Tyan DB, Chen S, Kaufman BD, Bernstein D, Rosenthal DN, Hollander SA. HLA desensitization with bortezomib in a highly sensitized pediatric patient. Pediatr Transplant 2014; 18:E280-2. [PMID: 25174602 DOI: 10.1111/petr.12347] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/30/2014] [Indexed: 11/28/2022]
Abstract
The proteasome inhibitor bortezomib has been used with variable success in the treatment of AMR following heart transplant. There is limited experience with this agent as a pretransplant desensitizing therapy. We report a case of successful HLA desensitization with a bortezomib-based protocol prior to successful heart transplantation. A nine-yr-old boy with dilated cardiomyopathy, not initially sensitized to HLA (cPRA of zero), required three days of ECMO, followed by implantation of a Heartmate II LVAD. Within six wk, the patient developed de novo class I IgG and C1q complement-fixing HLA antibodies with a cPRA of 100%. Two doses of IVIG (2 g/kg) failed to reduce antibody levels, although two courses of a novel desensitization protocol consisting of rituximab (375 mg/m(2) ), bortezomib (1.3 mg/m(2) × 5 doses), and plasmapheresis reduced his cPRA to 0% and 87% by the C1q and IgG assays, respectively. He underwent heart transplantation nearly two months later. The patient is now >one yr post-transplant, is free of both AMR and ACR, and has no detectable donor-specific antibodies by IgG or C1q. Proteasome inhibition with bortezomib and plasmapheresis may be an effective therapy for HLA desensitization pretransplant.
Collapse
Affiliation(s)
- Lindsay J May
- Division of Pediatric Cardiology, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Herrera AF, Kim HT, Bindra B, Jones KT, Alyea EP, Armand P, Cutler CS, Ho VT, Nikiforow S, Blazar BR, Ritz J, Antin JH, Soiffer RJ, Koreth J. A phase II study of bortezomib plus prednisone for initial therapy of chronic graft-versus-host disease. Biol Blood Marrow Transplant 2014; 20:1737-43. [PMID: 25017765 DOI: 10.1016/j.bbmt.2014.06.040] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 06/30/2014] [Indexed: 12/21/2022]
Abstract
Chronic graft-versus-host disease (GVHD) induces significant morbidity and mortality after allogeneic hematopoietic stem cell transplantation. Corticosteroids are standard initial therapy, despite limited efficacy and long-term toxicity. Based on our experience using bortezomib as effective acute GVHD prophylaxis, we hypothesized that proteasome-inhibition would complement the immunomodulatory effects of corticosteroids to improve outcomes in chronic GVHD (cGVHD). We undertook a single-arm phase II trial of bortezomib plus prednisone for initial therapy of cGVHD. Bortezomib was administered at 1.3 mg/m(2) i.v. on days 1, 8, 15, and 22 of each 35-day cycle for 3 cycles (15 weeks). Prednisone was dosed at .5 to 1 mg/kg/day, with a suggested taper after cycle 1. All 22 enrolled participants were evaluable for toxicity; 20 were evaluable for response. Bortezomib plus prednisone therapy was well tolerated, with 1 occurrence of grade 3 sensory peripheral neuropathy possibly related to bortezomib. The overall response rate at week 15 in evaluable participants was 80%, including 2 (10%) complete and 14 (70%) partial responses. The organ-specific complete response rate was 73% for skin, 53% for liver, 75% for gastrointestinal tract, and 33% for joint, muscle, or fascia involvement. The median prednisone dose decreased from 50 mg/day to 20 mg/day at week 15 (P < .001). The combination of bortezomib and prednisone for initial treatment of cGVHD is feasible and well tolerated. We observed a high response rate to combined bortezomib and prednisone therapy; however, in this single-arm study, we could not directly measure the impact of bortezomib. Proteasome inhibition may offer benefit in the treatment of cGVHD and should be further evaluated.
Collapse
Affiliation(s)
- Alex F Herrera
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Haesook T Kim
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Bhavjot Bindra
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kyle T Jones
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Edwin P Alyea
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Philippe Armand
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Corey S Cutler
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Vincent T Ho
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sarah Nikiforow
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Bruce R Blazar
- Blood and Marrow Transplantation, University of Minnesota Masonic Cancer Center, Minneapolis, Minnesota
| | - Jerome Ritz
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Joseph H Antin
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Robert J Soiffer
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - John Koreth
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts.
| |
Collapse
|
21
|
Gomez AM, Willcox N, Vrolix K, Hummel J, Nogales-Gadea G, Saxena A, Duimel H, Verheyen F, Molenaar PC, Buurman WA, De Baets MH, Martinez-Martinez P, Losen M. Proteasome inhibition with bortezomib depletes plasma cells and specific autoantibody production in primary thymic cell cultures from early-onset myasthenia gravis patients. THE JOURNAL OF IMMUNOLOGY 2014; 193:1055-1063. [PMID: 24973445 DOI: 10.4049/jimmunol.1301555] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Bortezomib is a potent inhibitor of proteasomes currently used to eliminate malignant plasma cells in multiple myeloma patients. It is also effective in depleting both alloreactive plasma cells in acute Ab-mediated transplant rejection and their autoreactive counterparts in animal models of lupus and myasthenia gravis (MG). In this study, we demonstrate that bortezomib at 10 nM or higher concentrations killed long-lived plasma cells in cultured thymus cells from nine early-onset MG patients and consistently halted their spontaneous production not only of autoantibodies against the acetylcholine receptor but also of total IgG. Surprisingly, lenalidomide and dexamethasone had little effect on plasma cells. After bortezomib treatment, they showed ultrastructural changes characteristic of endoplasmic reticulum stress after 8 h and were no longer detectable at 24 h. Bortezomib therefore appears promising for treating MG and possibly other Ab-mediated autoimmune or allergic disorders, especially when given in short courses at modest doses before the standard immunosuppressive drugs have taken effect.
Collapse
Affiliation(s)
- Alejandro M Gomez
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Nick Willcox
- Department of Clinical Neurology, University of Oxford, UK
| | - Kathleen Vrolix
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Jonas Hummel
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Gisela Nogales-Gadea
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,Neuromuscular Diseases Unit, Institut de Recerca del Hospital de la Santa Creu i Sant Pau, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Abhishek Saxena
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Hans Duimel
- ELMI Unit-CRISP, Department of Molecular Cell Biology, Maastricht University, Maastricht, the Netherlands
| | - Fons Verheyen
- ELMI Unit-CRISP, Department of Molecular Cell Biology, Maastricht University, Maastricht, the Netherlands
| | - Peter C Molenaar
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Wim A Buurman
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Marc H De Baets
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Pilar Martinez-Martinez
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Mario Losen
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
22
|
Abstract
Following infections and environmental exposures, memory T cells are generated that provide long-term protective immunity. Compared to their naïve T cell counterparts, memory T cells possess unique characteristics that endow them with the ability to quickly and robustly respond to foreign antigens. While such memory T cells are beneficial in protecting their hosts from recurrent infection, memory cells reactive to donor antigens pose a major barrier to successful transplantation and tolerance induction. Significant progress has been made over the past several decades contributing to our understanding of memory T cell generation, their distinct biology, and their detrimental impact in clinical and animal models of transplantation. This review focuses on the unique features which make memory T cells relevant to the transplant community and discusses potential therapies targeting memory T cells which may ameliorate allograft rejection.
Collapse
Affiliation(s)
- Charles A Su
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106 ; Glickman Urological and Kidney Institute and Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Robert L Fairchild
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106 ; Glickman Urological and Kidney Institute and Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195
| |
Collapse
|
23
|
Ezzelarab M, Zahorchak A, Lu L, Morelli A, Chalasani G, Demetris A, Lakkis F, Wijkstrom M, Murase N, Humar A, Shapiro R, Cooper D, Thomson A. Regulatory dendritic cell infusion prolongs kidney allograft survival in nonhuman primates. Am J Transplant 2013; 13:1989-2005. [PMID: 23758811 PMCID: PMC4070451 DOI: 10.1111/ajt.12310] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 04/03/2013] [Accepted: 04/16/2013] [Indexed: 01/25/2023]
Abstract
We examined the influence of regulatory dendritic cells (DCreg), generated from cytokine-mobilized donor blood monocytes in vitamin D3 and IL-10, on renal allograft survival in a clinically relevant rhesus macaque model. DCreg expressed low MHC class II and costimulatory molecules, but comparatively high levels of programmed death ligand-1 (B7-H1), and were resistant to pro-inflammatory cytokine-induced maturation. They were infused intravenously (3.5-10 × 10(6) /kg), together with the B7-CD28 costimulation blocking agent CTLA4Ig, 7 days before renal transplantation. CTLA4Ig was given for up to 8 weeks and rapamycin, started on Day -2, was maintained with tapering of blood levels until full withdrawal at 6 months. Median graft survival time was 39.5 days in control monkeys (no DC infusion; n = 6) and 113.5 days (p < 0.05) in DCreg-treated animals (n = 6). No adverse events were associated with DCreg infusion, and there was no evidence of induction of host sensitization based on circulating donor-specific alloantibody levels. Immunologic monitoring also revealed regulation of donor-reactive memory CD95(+) T cells and reduced memory/regulatory T cell ratios in DCreg-treated monkeys compared with controls. Termination allograft histology showed moderate combined T cell- and Ab-mediated rejection in both groups. These findings justify further preclinical evaluation of DCreg therapy and their therapeutic potential in organ transplantation.
Collapse
Affiliation(s)
- M. Ezzelarab
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine
| | - A.F. Zahorchak
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine
| | - L. Lu
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine
| | - A.E. Morelli
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - G. Chalasani
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine,Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - A.J. Demetris
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - F.G. Lakkis
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - M. Wijkstrom
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine
| | - N. Murase
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine
| | - A. Humar
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine
| | - R. Shapiro
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine
| | - D.K.C. Cooper
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine
| | - A.W. Thomson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA,Corresponding author: Angus W. Thomson, PhD DSc, University of Pittsburgh School of Medicine, 200 Lothrop Street, W1540 BST, Pittsburgh, PA 15261, Phone: (412) 624-6392,
| |
Collapse
|
24
|
Ramsey H, Pilat N, Hock K, Klaus C, Unger L, Schwarz C, Baranyi U, Gattringer M, Schwaiger E, Wrba F, Wekerle T. Anti-LFA-1 or rapamycin overcome costimulation blockade-resistant rejection in sensitized bone marrow recipients. Transpl Int 2012; 26:206-18. [PMID: 23240587 DOI: 10.1111/tri.12021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Revised: 01/15/2012] [Accepted: 10/25/2012] [Indexed: 02/04/2023]
Abstract
While costimulation blockade-based mixed chimerism protocols work well for inducing tolerance in rodents, translation to preclinical large animal/nonhuman primate models has been less successful. One recognized cause for these difficulties is the high frequency of alloreactive memory T cells (Tmem) found in the (pre)clinical setting as opposed to laboratory mice. In the present study, we therefore developed a murine bone marrow transplantation (BMT) model employing recipients harboring polyclonal donor-reactive Tmem without concomitant humoral sensitization. This model was then used to identify strategies to overcome this additional immune barrier. We found that B6 recipients that were enriched with 3 × 10(7) T cells isolated from B6 mice that had been previously grafted with Balb/c skin, rejected Balb/c BM despite costimulation blockade with anti-CD40L and CTLA4Ig (while recipients not enriched developed chimerism). Adjunctive short-term treatment of sensitized BMT recipients with rapamycin or anti-LFA-1 mAb was demonstrated to be effective in controlling Tmem in this model, leading to long-term mixed chimerism and donor-specific tolerance. Thus, rapamycin and anti-LFA-1 mAb are effective in overcoming the potent barrier that donor-reactive Tmem pose to the induction of mixed chimerism and tolerance despite costimulation blockade.
Collapse
Affiliation(s)
- Haley Ramsey
- Division of Transplantation, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Murine anti-third-party central-memory CD8(+) T cells promote hematopoietic chimerism under mild conditioning: lymph-node sequestration and deletion of anti-donor T cells. Blood 2012; 121:1220-8. [PMID: 23223359 DOI: 10.1182/blood-2012-07-441493] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Transplantation of T cell-depleted BM (TDBM) under mild conditioning, associated with minimal toxicity and reduced risk of GVHD, offers an attractive therapeutic option for patients with nonmalignant hematologic disorders and can mediate immune tolerance to subsequent organ transplantation. However, overcoming TDBM rejection after reduced conditioning remains a challenge. Here, we address this barrier using donorderived central memory CD8(+) T cells (Tcms), directed against third-party antigens. Our results show that fully allogeneic or (hostXdonor)F1-Tcm, support donor chimerism (> 6 months) in sublethally irradiated (5.5Gy) mice, without GVHD symptoms. Chimerism under yet lower irradiation (4.5Gy) was achieved by combining Tcm with short-term administration of low-dose Rapamycin. Importantly, this chimerism resulted in successful donor skin acceptance, whereas third-party skin was rejected. Tracking of host anti-donor T cells (HADTCs), that mediate TDBMT rejection, in a novel bioluminescence-imaging model revealed that Tcms both induce accumulation and eradicate HADTCs in the LNs,concomitant with their elimination from other organs, including the BM. Further analysis with 2-photon microcopy revealed that Tcms form conjugates with HADTCs, resulting in decelerated and confined movement of HADTCs within the LNs in an antigen-specific manner. Thus, anti-third-party Tcms support TDBMT engraftment under reduced-conditioning through lymph-node sequestration and deletion of HADTCs, offering a novel and potentially safe approach for attaining stable hematopoietic chimerism.
Collapse
|
26
|
Luo L, Sun Z, Cheng H, Luo G. Memory T-cell-specific therapeutics attenuate allograft rejection via mediation of alloreactivity in memory cells. Immunol Lett 2012; 148:53-8. [PMID: 22902239 DOI: 10.1016/j.imlet.2012.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 07/30/2012] [Accepted: 08/01/2012] [Indexed: 02/01/2023]
Abstract
Many means in inbred rodent models promoted long-term graft survival or donor-specific tolerance, but less so in nonhuman primates, outbred rodents or human patients. A diverse repertoire of memory T cells, derived from heterologous immunity or prior to exposure to alloantigen, has been believed to be an important part of this barrier. Memory T cells have a unique capacity to generate effector functions quickly upon re-exposure to antigen, and this capacity is achieved by reduced activation thresholds, and expressed high level trafficking and adhesion molecules, which is likely responsible for their exhibiting differential susceptibility to immune therapeutics compared with naïve T cells. This review outlines recent progress on characteristics of memory T cells and focuses on these potential therapies targeting memory T cells which are likely to ameliorate allograft rejection by inducing transplant tolerance.
Collapse
Affiliation(s)
- Lei Luo
- Department of Research and Education, Guizhou Province People's Hospital, Guiyang, China
| | | | | | | |
Collapse
|
27
|
Koreth J, Stevenson KE, Kim HT, McDonough SM, Bindra B, Armand P, Ho VT, Cutler C, Blazar BR, Antin JH, Soiffer RJ, Ritz J, Alyea EP. Bortezomib-based graft-versus-host disease prophylaxis in HLA-mismatched unrelated donor transplantation. J Clin Oncol 2012; 30:3202-8. [PMID: 22869883 DOI: 10.1200/jco.2012.42.0984] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE HLA-mismatched unrelated donor (MMUD) hematopoietic stem-cell transplantation (HSCT) is associated with increased graft-versus-host disease (GVHD) and impaired survival. In reduced-intensity conditioning (RIC), neither ex vivo nor in vivo T-cell depletion (eg, antithymocyte globulin) convincingly improved outcomes. The proteasome inhibitor bortezomib has immunomodulatory properties potentially beneficial for control of GVHD in T-cell-replete HLA-mismatched transplantation. PATIENTS AND METHODS We conducted a prospective phase I/II trial of a GVHD prophylaxis regimen of short-course bortezomib, administered once per day on days +1, +4, and +7 after peripheral blood stem-cell infusion plus standard tacrolimus and methotrexate in patients with hematologic malignancies undergoing MMUD RIC HSCT. We report outcomes for 45 study patients: 40 (89%) 1-locus and five (11%) 2-loci mismatches (HLA-A, -B, -C, -DRB1, or -DQB1), with a median of 36.5 months (range, 17.4 to 59.6 months) follow-up. RESULTS The 180-day cumulative incidence of grade 2 to 4 acute GVHD was 22% (95% CI, 11% to 35%). One-year cumulative incidence of chronic GVHD was 29% (95% CI, 16% to 43%). Two-year cumulative incidences of nonrelapse mortality (NRM) and relapse were 11% (95% CI, 4% to 22%) and 38% (95% CI, 24% to 52%), respectively. Two-year progression-free survival and overall survival were 51% (95% CI, 36% to 64%) and 64% (95% CI, 49% to 76%), respectively. Bortezomib-treated HLA-mismatched patients experienced rates of NRM, acute and chronic GVHD, and survival similar to those of contemporaneous HLA-matched RIC HSCT at our institution. Immune recovery, including CD8(+) T-cell and natural killer cell reconstitution, was enhanced with bortezomib. CONCLUSION A novel short-course, bortezomib-based GVHD regimen can abrogate the survival impairment of MMUD RIC HSCT, can enhance early immune reconstitution, and appears to be suitable for prospective randomized evaluation.
Collapse
Affiliation(s)
- John Koreth
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA 02215, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Caballero-Velázquez T, Sánchez-Abarca LI, Gutierrez-Cosio S, Blanco B, Calderon C, Herrero C, Carrancio S, Serrano C, del Cañizo C, San Miguel JF, Pérez-Simón JA. The novel combination of sirolimus and bortezomib prevents graft-versus-host disease but maintains the graft-versus-leukemia effect after allogeneic transplantation. Haematologica 2012; 97:1329-37. [PMID: 22532520 DOI: 10.3324/haematol.2011.058677] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND We have previously shown that bortezomib induces a depletion of alloreactive T cells and allows the expansion of T cells with suppressive properties. In the current study, we analyzed the potential synergistic effect of bortezomib in conjunction with sirolimus in order to reduce-graft-versus-host disease without hampering graft-versus-leukemia effect in the allogeneic transplant setting. DESIGN AND METHODS We evaluated the effect of sirolimus, bortezomib or the combination of both in the proliferation and activation of in vitro stimulated T lymphocytes. Pathways involved in this synergy were also analyzed using Western blot assays. Finally, BALB/c mice receiving C57BL/6 allogeneic donor bone marrow with splenocytes were used to measure in vivo the effect of this novel combination on the risk of graft-versus-host disease. RESULTS The combination of both drugs synergistically inhibited both activation and proliferation of stimulated T cells. Also, the production of Th1 cytokines (IFN γ, IL-2 and TNF) was significantly inhibited. This effect was due, at least in part, to the inhibition of Erk and Akt phosphorylation. In vivo, the combination reduced the risk of graft-versus-host disease without hampering graft-versus-leukemia effect, as shown in mice receiving graft-versus-host disease prophylaxis with sirolimus plus bortezomib being infused with tumor WEHI cells plus C57BL/6 donor BM and splenocytes. CONCLUSIONS The current study reveals a synergistic effect of the combination sirolimus and bortezomib to prevent graft-versus-host disease while maintaining the graft-versus-leukemia effect.
Collapse
Affiliation(s)
- Teresa Caballero-Velázquez
- Servicio de Hematología, Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Instituto de Biomedicina de Sevilla (IBIS), Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Harnessing regulatory T cells for transplant tolerance in the clinic through mTOR inhibition: myth or reality? Curr Opin Organ Transplant 2012; 16:606-13. [PMID: 21971509 DOI: 10.1097/mot.0b013e32834c237a] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE OF REVIEW The inhibition of mTOR promotes immune tolerance in mouse models of transplantation, by favoring the expansion of regulatory T cells over effector T cells. However, attempts at inducing immune tolerance with the mTOR inhibitor (mTOR-I) in humans have so far failed. We herein review the immunological obstacles that need to be overcome in order to translate mTOR-I-related tolerogenic properties into the clinic. RECENT FINDINGS Our knowledge of regulatory T-cell biology has exploded over the past few years, providing clues to explain the complex impact of prolonged mTOR inhibition on the biology of regulatory T cells. Furthermore, recent data have shed light on the unexpected pro-inflammatory burst observed in some transplant recipients treated with mTOR-I. We propose that the exposure of an organism to pathogens determines the immunodominant effect of mTOR-I, altering the immune system from a state of tolerance in inbred animals to a state of infection-triggered enhanced inflammation in humans. SUMMARY Recent advances in the understanding of the pleiotropic effects of mTOR-I on the immune system are paving the way to new therapeutic avenues. Future mTOR-I-based tolerogenic protocols should counter the mTOR-I-related inflammation in order to selectively promote expansion of stable regulatory T cells. We herein envisage promising therapeutic perspectives.
Collapse
|
30
|
Krummey SM, Ford ML. Heterogeneity within T Cell Memory: Implications for Transplant Tolerance. Front Immunol 2012; 3:36. [PMID: 22566919 PMCID: PMC3342058 DOI: 10.3389/fimmu.2012.00036] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 02/15/2012] [Indexed: 12/16/2022] Open
Abstract
Adaptive immunity in both mouse and man results in the generation of immunological memory. Memory T cells are both friend and foe to transplant recipients, as they are intimately involved and in many cases absolutely required for the maintenance of protective immunity in the face immunosuppression, yet from the evidence presented herein they clearly constitute a formidable barrier for the successful implementation of tolerance induction strategies in transplantation. This review describes the experimental evidence demonstrating the increased resistance of memory T cells to many distinct tolerance induction strategies, and outlines recent advances in our knowledge of the ways in which alloreactive memory T cells arise in previously untransplanted individuals. Understanding the impact of alloreactive memory T cell specificity, frequency, and quality might allow for better donor selection in order to minimize the donor-reactive memory T cell barrier in an individual transplant recipient, thus allowing stratification of relative risk of alloreactive memory T cell mediated rejection, and conversely increase the likelihood of successful establishment of tolerance. However, further research into the molecular and cellular pathways involved in alloreactive memory T cell-mediated rejection is required in order to design new strategies to overcome the memory T cell barrier, without critically impairing protective immunity.
Collapse
Affiliation(s)
- Scott M Krummey
- Department of Surgery, Emory Transplant Center, Emory University Atlanta, GA, USA
| | | |
Collapse
|
31
|
Kwun J, Bulut P, Kim E, Dar W, Oh B, Ruhil R, Iwakoshi N, Knechtle SJ. The role of B cells in solid organ transplantation. Semin Immunol 2011; 24:96-108. [PMID: 22137187 DOI: 10.1016/j.smim.2011.08.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 08/30/2011] [Indexed: 12/30/2022]
Abstract
The role of antibodies in chronic injury to organ transplants has been suggested for many years, but recently emphasized by new data. We have observed that when immunosuppressive potency decreases either by intentional weaning of maintenance agents or due to homeostatic repopulation after immune cell depletion, the threshold of B cell activation may be lowered. In human transplant recipients the result may be donor-specific antibody, C4d+ injury, and chronic rejection. This scenario has precise parallels in a rhesus monkey renal allograft model in which T cells are depleted with CD3 immunotoxin, or in a CD52-T cell transgenic mouse model using alemtuzumab to deplete T cells. Such animal models may be useful for the testing of therapeutic strategies to prevent DSA. We agree with others who suggest that weaning of immunosuppression may place transplant recipients at risk of chronic antibody-mediated rejection, and that strategies to prevent this scenario are needed if we are to improve long-term graft and patient outcomes in transplantation. We believe that animal models will play a crucial role in defining the pathophysiology of antibody-mediated rejection and in developing effective therapies to prevent graft injury. Two such animal models are described herein.
Collapse
Affiliation(s)
- Jean Kwun
- Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Bartel G, Schwaiger E, Böhmig GA. Prevention and treatment of alloantibody-mediated kidney transplant rejection. Transpl Int 2011; 24:1142-55. [PMID: 21831227 DOI: 10.1111/j.1432-2277.2011.01309.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Antibody-mediated rejection (AMR), which is commonly caused by preformed and/or de novo HLA alloantibodies, has evolved as a leading cause of early and late kidney allograft injury. In recent years, effective treatment strategies have been established to counteract the deleterious effects of humoral alloreactivity. One major therapeutic challenge is the barrier of a positive pretransplant lymphocytotoxic crossmatch. Several apheresis- and/or IVIG-based protocols have been shown to enable successful crossmatch conversion, including a strategy of peritransplant immunoadsorption for rapid crossmatch conversion immediately before deceased donor transplantation. While such protocols may increase transplant rates and allow for acceptable graft survival, at least in the short-term, it has become evident that, despite intense treatment, many patients still experience clinical or subclinical AMR. This reinforces the need for innovative strategies, such as complementary allocation programs to improve transplant outcomes. For acute AMR, various studies have suggested efficiency of plasmapheresis- or immunoadsorption-based protocols. There is, however, no established treatment for chronic AMR and the development of strategies to reverse or at least halt chronic active rejection remains a big challenge. Major improvements can be expected from studies evaluating innovative therapeutic concepts, such as proteasome inhibition or complement blocking agents.
Collapse
Affiliation(s)
- Gregor Bartel
- Department of Medicine III, Medical University Vienna, Vienna, Austria
| | | | | |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW Advances in surgical techniques and combinations of conventional immunosuppressants have made paediatric liver transplantation the success story it is today. However, the increasing numbers of survivors reaching adulthood highlight important issues of long-term quality of life and drug induced complications. The aim of this review is to describe the trends and advances in immunosuppression for paediatric liver transplantation over the last 12 months. RECENT DEVELOPMENTS As our knowledge of the immune cell populations and intracellular mechanisms involved in alloreactivity improves, induction immunosuppression has emerged as a powerful therapeutic manoeuvre to counter the initial alloimmune response. Many centres have adopted a more focused use of biological agents at induction to improve immunosuppression in the critical peritransplant period and to reduce the level of subsequent maintenance requirements. Corticosteroid avoidance and calcineurin inhibitors minimization trials have obtained encouraging results. New immunosuppressive strategies have evolved towards the goal of inducing operational tolerance, and paediatric liver transplant recipients seem to be a particularly promising target. New strategies are being developed also to improve quality of life and reduce nonadherence in adolescents and young adults who underwent liver transplantation. New drugs target B-cell and complement driven rejection and new monoclonal antibodies and small molecules are under trial to inhibit specific signals in the immune response. SUMMARY We review current trends of immunosuppressive protocols in paediatric liver transplantation, focusing on induction agents, corticosteroid avoidance and calcineurin inhibitor sparing protocols, protocols for adult transition and new drugs currently under development.
Collapse
|
34
|
Non-human primate regulatory T cells: current biology and implications for transplantation. Transplantation 2010; 90:811-6. [PMID: 20671597 DOI: 10.1097/tp.0b013e3181ebf782] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Regulatory T cells (Treg) offer potential for improving long-term outcomes in cell and organ transplantation. The non-human primate model is a valuable resource for addressing issues concerning the transfer of Treg therapy to the clinic. Herein, we discuss the properties of non-human primate Treg and prospects for their evaluation in allotransplantation and xenotransplantation.
Collapse
|
35
|
Overcoming the memory barrier in tolerance induction: molecular mimicry and functional heterogeneity among pathogen-specific T-cell populations. Curr Opin Organ Transplant 2010; 15:405-10. [PMID: 20616729 DOI: 10.1097/mot.0b013e32833b7916] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE OF REVIEW This review highlights recent advances in our understanding of the frequency and nature of alloreactivity among memory T-cell populations, and discusses recent successes in experimentally targeting these populations in order to prolong graft survival. RECENT FINDINGS Recent studies suggest that not only is alloreactivity present within peripheral T-cell compartments of normal healthy individuals, but cross-reactivity between viral-specific T cells and allotropes may in fact be a very common occurrence. Furthermore, this cross-reactivity functions at the level of molecular mimicry of T-cell receptor recognition. Therapeutics that specifically target cell surface molecules or effector pathways used by memory T cells to mediate graft rejection will likely be required in order to attenuate the donor-reactive memory T-cell response during transplantation. SUMMARY A major challenge facing the field over the next decade is to define the heterogeneity that exists within memory T-cell populations that impacts graft survival. Understanding the functional and phenotypic differences that modify the memory T-cell barrier to tolerance induction might allow a strategy in which strength of immunosuppression could be tailored to fit the immunological history of a given transplant recipient in order to minimize nonimmune toxicities, maximize protective immunity, and prolong graft survival.
Collapse
|
36
|
Lemy A, Toungouz M, Abramowicz D. Bortezomib: a new player in pre- and post-transplant desensitization? Nephrol Dial Transplant 2010; 25:3480-9. [PMID: 20826741 DOI: 10.1093/ndt/gfq502] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Several desensitization strategies have been investigated for the reversal of acute antibody-mediated rejection or for the removal of preformed anti-HLA antibodies, with the aim to promote access to renal transplantation. Today, their success appears limited or incomplete. Bortezomib, a selective inhibitor of the 26S proteasome, which is largely used in the treatment of multiple myeloma, could be a novel promising desensitizing agent. Its mechanism of action and preliminary clinical use in renal transplantation is reviewed here.
Collapse
|