1
|
Oxley EP, Kershaw NJ, Louis C, Goodall KJ, Garwood MM, Jee Ho SM, Voo VTF, Park HY, Iaria J, Wong LLL, Lebenbaum AG, Wiranata S, Pang ES, Edwards ESJ, D'Silva DB, Hansen J, van Zelm MC, O'Keeffe M, Hogarth PM, Haynes NM, Huntington ND, Wicks IP, Dickins RA. Context-restricted PD-(L)1 checkpoint agonism by CTLA4-Ig therapies inhibits T cell activity. Cell Rep 2024; 43:114834. [PMID: 39383033 DOI: 10.1016/j.celrep.2024.114834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/30/2024] [Accepted: 09/19/2024] [Indexed: 10/11/2024] Open
Abstract
T cell surface CTLA4 sequesters the costimulatory ligands CD80 and CD86 on antigen-presenting cells (APCs) to prevent autoimmunity. Therapeutic immunosuppression by recombinant CTLA4-immunoglobulin (Ig) fusion proteins, including abatacept, is also attributed to CD80/CD86 blockade. Recent studies show that CTLA4-Ig binding to APC surface cis-CD80:PD-L1 complexes can release the inhibitory ligand PD-L1, but whether this contributes to T cell inhibition remains unclear. Here, we show that PD-L1 liberation by CTLA4-Ig is strictly limited, both in extent and context, relative to PD-L1-competing anti-CD80 antibodies. At APC surface CD80:PD-L1 ratios exceeding 2:1, CTLA4-Ig therapies fail to release PD-L1 regardless of their CD80 affinity. Additionally, introducing flexibility into CTLA4-Ig by modifying its rigid homodimer interface produces biologics that retain bivalent CD80 binding without dissociating cis-bound PD-L1. These findings demonstrate that CTLA4-Ig therapies liberate PD-L1 through a CD80 reorientation mechanism that imposes a strict context dependence to their PD-1 checkpoint agonism and resultant T cell inhibition.
Collapse
Affiliation(s)
- Ethan P Oxley
- Australian Centre for Blood Diseases, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Nadia J Kershaw
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Royal Parade, Parkville, VIC 3052, Australia
| | - Cynthia Louis
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Royal Parade, Parkville, VIC 3052, Australia
| | - Katharine J Goodall
- Australian Centre for Blood Diseases, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Maximilian M Garwood
- Australian Centre for Blood Diseases, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Skye Min Jee Ho
- Australian Centre for Blood Diseases, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Veronica T F Voo
- Australian Centre for Blood Diseases, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Hae-Young Park
- Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Josephine Iaria
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia
| | - Lilian L L Wong
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia
| | - Ariel G Lebenbaum
- Australian Centre for Blood Diseases, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Stephanie Wiranata
- Australian Centre for Blood Diseases, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Ee Shan Pang
- Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Emily S J Edwards
- Department of Immunology and Pathology, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Damian B D'Silva
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia
| | - Jacinta Hansen
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia
| | - Menno C van Zelm
- Department of Immunology and Pathology, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia; Department of Allergy, Immunology & Respiratory Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Meredith O'Keeffe
- Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - P Mark Hogarth
- Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia; Department of Clinical Pathology, The University of Melbourne, Royal Parade, Parkville, VIC 3052, Australia
| | - Nicole M Haynes
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville VIC 3052, Australia
| | - Nicholas D Huntington
- Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Ian P Wicks
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Royal Parade, Parkville, VIC 3052, Australia
| | - Ross A Dickins
- Australian Centre for Blood Diseases, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia.
| |
Collapse
|
2
|
Ortiz V, Loeuillard E. Rethinking Immune Check Point Inhibitors Use in Liver Transplantation: Implications and Resistance. Cell Mol Gastroenterol Hepatol 2024; 19:101407. [PMID: 39326581 PMCID: PMC11609388 DOI: 10.1016/j.jcmgh.2024.101407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy, including the two most common liver tumors, hepatocellular carcinoma and cholangiocarcinoma, but their use in the peri-transplantation period is controversial. ICI therapy aims to heighten cytotoxic T lymphocytes response against tumors. However, tumor recurrence is common owing to tumor immune response escape involving ablation of CTL response by interfering with antigen presentation, triggering CLT apoptosis and inducing epigenetic changes that promote ICI therapy resistance. ICI can also affect tissue resident memory T cell population, impact tolerance in the post-transplant period, and induce acute inflammation risking graft survival post-transplant. Their interaction with immunosuppression may be key in reducing tumor burden and may thus, require multimodal therapy to treat these tumors. This review summarizes ICI use in the liver transplantation period, their impact on tolerance and resistance, and new potential therapies for combination or sequential treatments for liver tumors.
Collapse
Affiliation(s)
- Vivian Ortiz
- Division of Gastroenterology, Department of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, Missouri.
| | | |
Collapse
|
3
|
Zhou X, Xu Q, Li W, Dong N, Stomberski C, Narla G, Lin Z. Protein Phosphatase 2A Activation Promotes Heart Transplant Acceptance in Mice. Transplantation 2024; 108:e36-e48. [PMID: 38126420 PMCID: PMC10922415 DOI: 10.1097/tp.0000000000004832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
BACKGROUND Although heart transplantation is the definitive treatment for heart failure in eligible patients, both acute and chronic transplant rejection frequently occur. Protein phosphatase 2A (PP2A) activity is critical in maintaining tissue and organ homeostasis. In this study, we evaluated the effect of a novel class of small molecule activators of PP2A (SMAPs) on allograft rejection in a mouse heterotopic heart transplantation model. METHODS Recipient mice were administered with DT-061 (a pharmaceutically optimized SMAP) or vehicle by oral gavage beginning 1 d after transplantation. Histological and immunofluorescence analyses were performed to examine allograft rejection. Regulatory T cells (Treg) from recipient spleens were subjected to flow cytometry and RNA sequencing analysis. Finally, the effect of DT-061 on smooth muscle cells (SMCs) migration and proliferation was assessed. RESULTS DT-061 treatment prolonged cardiac allograft survival. SMAPs effectively suppressed the inflammatory immune response while increasing Treg population in the allografts, findings corroborated by functional analysis of RNA sequencing data derived from Treg of treated splenic tissues. Importantly, SMAPs extended immunosuppressive agent cytotoxic T lymphocyte-associated antigen-4-Ig-induced cardiac transplantation tolerance and allograft survival. SMAPs also strongly mitigated cardiac allograft vasculopathy as evidenced by a marked reduction of neointimal hyperplasia and SMC proliferation. Finally, our in vitro studies implicate suppression of MEK/ERK pathways as a unifying mechanism for the effect of PP2A modulation in Treg and SMCs. CONCLUSIONS PP2A activation prevents cardiac rejection and prolongs allograft survival in a murine model. Our findings highlight the potential of PP2A activation in improving alloengraftment in heart transplantation.
Collapse
Affiliation(s)
- Xianming Zhou
- Cardiology Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Xu
- Cardiology Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Department of Cardiovascular Surgery, Xiangya Hospital of Central South University, Changsha, China
| | - Wangzi Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Colin Stomberski
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Goutham Narla
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Zhiyong Lin
- Cardiology Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
4
|
Chang Y, Xu M, Zhang Y, Chen X, Sheng Y, Tao M, Zhang H, Xu Z, Hu S, Song J. Ruxolitinib attenuates acute rejection and can serve as an immune induction therapy in heart transplantation. Clin Immunol 2023; 257:109851. [PMID: 38008145 DOI: 10.1016/j.clim.2023.109851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 10/21/2023] [Accepted: 11/09/2023] [Indexed: 11/28/2023]
Abstract
The benefits of IL2RA antagonists in heart transplant patients are controversial. We aimed to elucidate the effects of IL2RA antagonists and identify targets that could be better than IL2RA antagonists. By using single-cell RNA sequencing of immune cells at different time points in patients receiving IL2RA antagonists, we identified nineteen types of cells. We revealed higher IL2RA expression in regulatory T cells (Tregs), suggesting that IL2RA antagonists attenuated IL-2-induced Treg activation. CD4_C04_IFNGR1 and CD8_C05_IFITM2 which had more cytotoxic effects, remained elevated at later time points. IFNGR1 was upregulated in these two subtypes, but was not expressed in Treg. Ruxolitinib targeted the pathways of IFNGR1 (JAK1/2) while not affecting the pathway of IL-2-induced Tregs activation (JAK3). Ruxolitinib showed prolonged survival compared to IL2RA mAb-treated mice. Our study provided dynamic changes of immune cells after IL2RA antagonists treatment at single-cell resolution. Ruxolitinib has potential as a new immunoinduction therapy without affecting Treg.
Collapse
Affiliation(s)
- Yuan Chang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing 100037, China
| | - Mengda Xu
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing 100037, China
| | - Yu Zhang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing 100037, China
| | - Xiao Chen
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing 100037, China
| | - Yixuan Sheng
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing 100037, China
| | - Menghao Tao
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing 100037, China
| | - Hang Zhang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing 100037, China
| | - Zhenyu Xu
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing 100037, China
| | - Shengshou Hu
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing 100037, China; Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen 518057, China.
| | - Jiangping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing 100037, China; Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen 518057, China.
| |
Collapse
|
5
|
Aghbash PS, Rasizadeh R, Arefi V, Nahand JS, Baghi HB. Immune-checkpoint expression in antigen-presenting cells (APCs) of cytomegaloviruses infection after transplantation: as a diagnostic biomarker. Arch Microbiol 2023; 205:280. [PMID: 37430000 DOI: 10.1007/s00203-023-03623-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/26/2023] [Accepted: 06/30/2023] [Indexed: 07/12/2023]
Abstract
Cytomegalovirus (CMV), a member of the Herpesviridae family, mostly causes only slight feverish symptoms or can be asymptomatic in immunocompetent individuals. However, it is known to be particularly a significant cause of morbidity in immunocompromised patients, including transplant recipients, whose immune system has been weakened due to the consumption of immunosuppressor drugs. Therefore, the diagnosis of CMV infection after transplantation is crucial. New diagnostic methods for the quick detection of CMV have been developed as a result of understanding the clinical importance of invasive CMV. Antigen-presenting cells (APCs) and T cells are important components of the immune system and it may be possible to diagnose viral infections using immunological markers, such as lymphocytosis, cytotoxic T lymphocytes (CTL), and serum cytokine levels. Moreover, PD-1, CTLA 4, and TIGIT, which are expressed on certain T cells and antigen-presenting cells, are over-expressed during the infection. The assessment of CMV infection based on T cell and APC activity, and the expression of immunological checkpoints, can be helpful for the diagnosis of transplant patients at risk for CMV infection. In this review, we will investigate how immune checkpoints affect immune cells and how they impair organ transplantation after CMV infection.
Collapse
Affiliation(s)
- Parisa Shiri Aghbash
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reyhaneh Rasizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Arefi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 5166/15731, Iran
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 5166/15731, Iran
| | - Hossein Bannazadeh Baghi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 5166/15731, Iran.
| |
Collapse
|
6
|
Habib JG, Liu D, Crepeau RM, Wagener ME, Ford ML. Selective CD28 blockade impacts T cell differentiation during homeostatic reconstitution following lymphodepletion. Front Immunol 2023; 13:1081163. [PMID: 36761170 PMCID: PMC9904166 DOI: 10.3389/fimmu.2022.1081163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/28/2022] [Indexed: 01/26/2023] Open
Abstract
Introduction Costimulation blockade targeting the CD28 pathway provides improved long-term renal allograft survival compared to calcineurin inhibitors but may be limited as CTLA-4-Ig (abatacept, belatacept) blocks both CD28 costimulation and CTLA-4 coinhibition. Directly targeting CD28 while leaving CTLA-4 intact may provide a mechanistic advantage. Fc-silent non-crosslinking CD28 antagonizing domain antibodies (dAb) are currently in clinical trials for renal transplantation. Given the current standard of care in renal transplantation at most US centers, it is likely that lymphodepletion via thymoglobulin induction therapy could be used in patients treated with CD28 antagonists. Thus, we investigated the impact of T cell depletion (TCD) on T cell phenotype following homeostatic reconstitution in a murine model of skin transplantation treated with anti-CD28dAb. Methods Skin from BALB/cJ donors was grafted onto C56BL/6 recipients which were treated with or without 0.2mg anti-CD4 and 10μg anti-CD8 one day prior to transplant and with or without 100μg anti-CD28dAb on days 0, 2, 4, 6, and weekly thereafter. Mice were euthanized six weeks post-transplant and lymphoid cells were analyzed by flow cytometry. Results Anti-CD28dAb reversed lymphopenia-induced differentiation of memory CD4+ T cells in the spleen and lymph node compared to TCD alone. Mice treated with TCD+anti-CD28dAb exhibited significantly improved skin graft survival compared to anti-CD28dAb alone, which was also improved compared to no treatment. In addition, the expression of CD69 was reduced on CD4+ and CD8+ T cells in the spleen and lymph node from mice that received TCD+anti-CD28dAb compared to TCD alone. While a reduced frequency of CD4+FoxP3+ T cells was observed in anti-CD28dAb treated mice relative to untreated controls, this was balanced by an increased frequency of CD8+Foxp3+ T cells that was observed in the blood and kidney of mice given TCD+anti-CD28dAb compared to TCD alone. Discussion These data demonstrate that CD28 signaling impacts the differentiation of both CD4+ and CD8+ T cells during homeostatic reconstitution following lymphodepletion, resulting in a shift towards fewer activated memory T cells and more CD8+FoxP3+ T cells, a profile that may underpin the observed prolongation in allograft survival.
Collapse
|
7
|
Ravichandran R, Itabashi Y, Fleming T, Bansal S, Bowen S, Poulson C, Bharat A, Bremner R, Smith M, Mohanakumar T. Low-dose IL-2 prevents murine chronic cardiac allograft rejection: Role for IL-2-induced T regulatory cells and exosomes with PD-L1 and CD73. Am J Transplant 2022; 22:2180-2194. [PMID: 35603986 DOI: 10.1111/ajt.17101] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 05/12/2022] [Accepted: 05/12/2022] [Indexed: 01/25/2023]
Abstract
To determine the effects and immunological mechanisms of low-dose interleukin-2 (IL-2) in a murine model of chronic cardiac allograft rejection (BALB/c to C57BL/6) after costimulatory blockade consisting of MR1 (250 μg/ip day 0) and CTLA4-Ig (200 μg/ip day 2), we administered low-dose IL-2 (2000 IU/day) starting on posttransplant day 14 for 3 weeks. T regulatory (Treg) cell infiltration of the grafts was determined by immunohistochemistry; circulating exosomes by western blot and aldehyde bead flow cytometry; antibodies to donor MHC by immunofluorescent staining of donor cells; and antibodies to cardiac self-antigens (myosin, vimentin) by ELISA. We demonstrated that costimulation blockade after allogeneic heart transplantation induced circulating exosomes containing cardiac self-antigens and antibodies to both donor MHC and self-antigens, leading to chronic rejection by day 45. Treatment with low-dose IL-2 prolonged allograft survival (>100 days), prevented chronic rejection, and induced splenic and graft-infiltrating CD4+ CD25+ Foxp3 Treg cells by day 45 and circulating exosomes (Foxp3+) with PD-L1 and CD73. MicroRNA 142, associated with the TGFβ pathway, was significantly downregulated in exosomes from IL-2-treated mice. In conclusion, low-dose IL-2 delays rejection in a murine model of chronic cardiac allograft rejection and also induces graft-infiltrating Tregs and circulating exosomes with immunoregulatory molecules.
Collapse
Affiliation(s)
| | - Yoshihiro Itabashi
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Timothy Fleming
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Sandhya Bansal
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Sara Bowen
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Christin Poulson
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Ankit Bharat
- Department of surgery, Northwestern University, Chicago, Illinois, USA
| | - Ross Bremner
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Michael Smith
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | | |
Collapse
|
8
|
Pardinhas C, Leal R, Figueiredo C, Rodrigues L, Guedes M, Santos L, Romãozinho C, Sá H, Alves R, Figueiredo A. Kidney Retransplant: Not Too Old for a Second Chance. Transplant Proc 2022; 54:1242-1246. [DOI: 10.1016/j.transproceed.2022.04.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/22/2022] [Accepted: 04/02/2022] [Indexed: 11/16/2022]
|
9
|
Vitale G, Gitto S, Campani C, Turco L, Baldan A, Marra F, Morelli MC. Biological therapies in patients with liver disease: are they really lifesavers? Expert Opin Biol Ther 2021; 22:473-490. [PMID: 34860629 DOI: 10.1080/14712598.2022.2013799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION The liver plays a key role in the setting of immune tolerance. Targeting antigens for presentation by antigen-presenting cells in the liver can induce immune tolerance to either autoantigens from the liver itself or organs outside of the liver. Despite its non-conventional capacity for tolerance induction, the liver remains a target organ for autoimmune diseases. Whereas chronic inflammation and intra-hepatic immuno-suppressive microenvironment occurring during liver fibrosis lead to hepatocellular carcinoma. Monoclonal antibodies have revolutionized the therapeutic strategies of many autoimmune diseases and some cancers. AREAS COVERED We review data from literature regarding the safety and efficacy of biologics in treating hepatobiliary autoimmune diseases and primary liver cancers. Furthermore, we describe their potential use in the setting of liver transplants and their main immune-related liver adverse events. EXPERT OPINION Biological therapies have changed the natural history of main autoimmune diseases and solid cancers. Compared to other organs and disease settings, the liver lags behind in biologics and their applications. The development of novel diagnostic and therapeutic strategies based on the immunological and antigenic characteristics of the hepatobiliary system could reduce mortality and transplant rates linked to chronic liver diseases.
Collapse
Affiliation(s)
- Giovanni Vitale
- Division of Internal Medicine for the Treatment of Severe Organ Failure, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Stefano Gitto
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Claudia Campani
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Laura Turco
- Division of Internal Medicine for the Treatment of Severe Organ Failure, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Anna Baldan
- Division of Internal Medicine for the Treatment of Severe Organ Failure, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Fabio Marra
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Maria Cristina Morelli
- Division of Internal Medicine for the Treatment of Severe Organ Failure, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| |
Collapse
|
10
|
La Muraglia GM, Zeng S, Crichton ES, Wagener ME, Ford ML, Badell IR. Superior inhibition of alloantibody responses with selective CD28 blockade is CTLA-4 dependent and T follicular helper cell specific. Am J Transplant 2021; 21:73-86. [PMID: 32406182 PMCID: PMC7665991 DOI: 10.1111/ajt.16004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 04/15/2020] [Accepted: 05/07/2020] [Indexed: 01/25/2023]
Abstract
Anti-donor antibodies cause immunologic injury in transplantation. CD28 blockade with CTLA-4-Ig has the ability to reduce the incidence of these donor-specific antibodies (DSA), but its mechanism is suboptimal for the inhibition of alloimmunity in that CTLA-4-Ig blocks both CD28 costimulation and CTLA-4 coinhibition. Thus selective CD28 blockade that spares CTLA-4 has potential to result in improved inhibition of humoral alloimmunity. To test this possibility, we utilized a full allogeneic mismatch murine transplant model and T follicular helper (Tfh):B cell co-culture system. We observed that selective blockade with an anti-CD28 domain antibody (dAb) compared to CTLA-4-Ig led to superior inhibition of Tfh cell, germinal center, and DSA responses in vivo and better control of B cell responses in vitro. CTLA-4 blockade enhanced the humoral alloresponse and, in combination with anti-CD28 dAb, abrogated the effects of selective blockade. This CTLA-4-dependent inhibition was Tfh cell specific in that CTLA-4 expression by Tfh cells was necessary and sufficient for the improved humoral inhibition observed with selective CD28 blockade. As CD28 blockade attracts interest for control of alloantibodies in the clinic, these data support selective CD28 blockade as a superior strategy to address DSA via the sparing of CTLA-4 and more potent targeting of Tfh cells.
Collapse
Affiliation(s)
| | - Susan Zeng
- Emory Transplant Center, Atlanta, GA, USA
| | | | | | | | | |
Collapse
|
11
|
Al Jarroudi O, Ulusakarya A, Almohamad W, Afqir S, Morere JF. Anti-Programmed Cell Death Protein 1 (PD-1) Immunotherapy for Metastatic Hepatocellular Carcinoma After Liver Transplantation: A Report of Three Cases. Cureus 2020; 12:e11150. [PMID: 33133796 PMCID: PMC7586420 DOI: 10.7759/cureus.11150] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The treatment of recurrent hepatocellular carcinoma (HCC) after liver transplantation is difficult due to the lack of effective treatment options. The available evidence on the emerging immunotherapy in liver transplantation is based on anecdotal experiences and requires additional investigations. To determine the efficacy and safety of immunotherapy in liver transplant recipients, we report three cases of recurrent metastatic HCC after liver transplantation who were treated with nivolumab as off-label salvage therapy.
Collapse
Affiliation(s)
| | - Ayhan Ulusakarya
- Medical Oncology, Assistance Publique - Hôpitaux de Paris (AP-HP) Paul Brousse Hospital, Paris, FRA
| | - Wathek Almohamad
- Medical Oncology, Assistance Publique - Hôpitaux de Paris (AP-HP) Paul Brousse Hospital, Paris, FRA
| | - Said Afqir
- Medical Oncology, University Hospital Mohammed VI, Oujda, MAR
| | - Jean-Francois Morere
- Medical Oncology, Assistance Publique - Hôpitaux de Paris (AP-HP) Paul Brousse Hospital, Paris, FRA
| |
Collapse
|
12
|
Abstract
T-cell immunity undergoes a complex and continuous remodeling with aging. Understanding those dynamics is essential in refining immunosuppression. Aging is linked to phenotypic and metabolic changes in T-cell immunity, many resulting into impaired function and compromised effectiveness. Those changes may impact clinical immunosuppression with evidences suggesting age-specific efficacies of some (CNI and mammalian target of rapamycin inhibitors) but not necessarily all immunosuppressants. Metabolic changes of T cells with aging have only recently been appreciated and may provide novel ways of immunosuppression. Here, we provide an update on changes of T-cell immunity in aging.
Collapse
|
13
|
Escoin-Perez C, Blasco S, Juan-Vidal O. Immune checkpoint inhibitors in special populations. A focus on advanced lung cancer patients. Lung Cancer 2020; 144:1-9. [PMID: 32278215 DOI: 10.1016/j.lungcan.2020.03.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/25/2020] [Accepted: 03/29/2020] [Indexed: 12/13/2022]
Abstract
Immune checkpoint inhibitors (ICIs), including those targeting programmed cell death 1 (PD-1), its ligand 1 (PD-L1), or cytotoxic T-lymphocyte antigen 4 (CTLA-4) have become the standard treatment for several malignancies, including lung cancer. However, some patient populations have been routinely excluded from clinical trials or are underrepresented in these studies, as is the case of elderly patients or patients with poor performance status, brain metastases, solid organ transplant, autoimmune diseases, chronic viral infections (such as human immunodeficiency virus or chronic viral hepatitis B and C), or organ dysfunction. Thus, the safety and efficacy of ICIs in these special populations is still unclear, despite regulatory approval of these agents. This review analyzes and summarizes the available information on the efficacy and safety of ICIs in these special populations, focusing on patients with lung cancer.
Collapse
Affiliation(s)
- Corina Escoin-Perez
- Department of Medical Oncology, Hospital Universitario de La Ribera, Crta. Corbera, Km1. 46600 Alzira, Valencia, Spain.
| | - Sara Blasco
- Department of Medical Oncology, Hospital de Sagunto, Av. Ramón y Cajal, s/n. 46520 Sagunto, Valencia, Spain.
| | - Oscar Juan-Vidal
- Department of Medical Oncology, Hospital Universitario y Politécnico La Fe, Av. Fernando Abril Martorell, 106. 46026, Valencia, Spain.
| |
Collapse
|
14
|
Cortés-Hernández A, Alvarez-Salazar E, Arteaga-Cruz S, Alberu-Gómez J, Soldevila G. Ex vivo expansion of regulatory T cells from long-term Belatacept-treated kidney transplant patients restores their phenotype and suppressive function but not their FOXP3 TSDR demethylation status. Cell Immunol 2020; 348:104044. [PMID: 32005344 DOI: 10.1016/j.cellimm.2020.104044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/08/2020] [Accepted: 01/11/2020] [Indexed: 02/09/2023]
Abstract
We recently reported that Tregs from long-term Belatacept-treated kidney transplant patients displayed an altered phenotype and impaired suppressive function compared to Tregs from healthy controls. However, it remains unknown whether ex vivo expansion of Tregs from patients who underwent long-term immunosuppression may be feasible to be used in their treatment. In this work, Tregs from Belatacept-treated patients were polyclonally expanded in vitro in the presence of rapamycin and IL-2. After four weeks of expansion, Tregs from patients expressed high levels of FOXP3, CD25, CTLA-4, Helios and CCR7, and showed strong suppressive activity, even in the presence of pro-inflammatory cytokines. However, FOXP3 TSDR demethylation remained lower in expanded Tregs from Belatacept-treated patients compared to healthy control Tregs. These data suggest that ex vivo expansion of Tregs from patients undergoing long-term immunosuppression may require the use of epigenetic modifying agents to stabilize FOXP3 expression to be considered as treatment in kidney transplant patients.
Collapse
Affiliation(s)
- A Cortés-Hernández
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - E Alvarez-Salazar
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - S Arteaga-Cruz
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - J Alberu-Gómez
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey, N.L., México 64710, Mexico
| | - G Soldevila
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|
15
|
Yeung MY, Grimmig T, Sayegh MH. Costimulation Blockade in Transplantation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1189:267-312. [PMID: 31758538 DOI: 10.1007/978-981-32-9717-3_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
T cells play a pivotal role in orchestrating immune responses directed against a foreign (allogeneic) graft. For T cells to become fully activated, the T-cell receptor (TCR) must interact with the major histocompatibility complex (MHC) plus peptide complex on antigen-presenting cells (APCs), followed by a second "positive" costimulatory signal. In the absence of this second signal, T cells become anergic or undergo deletion. By blocking positive costimulatory signaling, T-cell allo-responses can be aborted, thus preventing graft rejection and promoting long-term allograft survival and possibly tolerance (Alegre ML, Najafian N, Curr Mol Med 6:843-857, 2006; Li XC, Rothstein DM, Sayegh MH, Immunol Rev 229:271-293, 2009). In addition, costimulatory molecules can provide negative "coinhibitory" signals that inhibit T-cell activation and terminate immune responses; strategies to promote these pathways can also lead to graft tolerance (Boenisch O, Sayegh MH, Najafian N, Curr Opin Organ Transplant 13:373-378, 2008). However, T-cell costimulation involves an incredibly complex array of interactions that may act simultaneously or at different times in the immune response and whose relative importance varies depending on the different T-cell subsets and activation status. In transplantation, the presence of foreign alloantigen incites not only destructive T effector cells but also protective regulatory T cells, the balance of which ultimately determines the fate of the allograft (Lechler RI, Garden OA, Turka LA, Nat Rev Immunol 3:147-158, 2003). Since the processes of alloantigen-specific rejection and regulation both require activation of T cells, costimulatory interactions may have opposing or synergistic roles depending on the cell being targeted. Such complexities present both challenges and opportunities in targeting T-cell costimulatory pathways for therapeutic purposes. In this chapter, we summarize our current knowledge of the various costimulatory pathways in transplantation and review the current state and challenges of harnessing these pathways to promote graft tolerance (summarized in Table 10.1).
Collapse
Affiliation(s)
- Melissa Y Yeung
- Department of Medicine, Renal Division, Brigham and Women's Hospital, Boston, MA, USA. .,Harvard Medical School, Boston, MA, USA.
| | - Tanja Grimmig
- Department of Surgery, Molecular Oncology and Immunology, University of Wuerzburg, Wuerzburg, Germany
| | - Mohamed H Sayegh
- Department of Medicine, Renal Division, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Department of Medicine and Immunology, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
16
|
Mardomi A, Mohammadi N, Khosroshahi HT, Abediankenari S. An update on potentials and promises of T cell co-signaling molecules in transplantation. J Cell Physiol 2019; 235:4183-4197. [PMID: 31696513 DOI: 10.1002/jcp.29369] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 10/07/2019] [Indexed: 02/06/2023]
Abstract
The promising outcomes of immune-checkpoint based immunotherapies in cancer have provided a proportional perspective ahead of exploiting similar approaches in allotransplantation. Belatacept (CTLA-4-Ig) is an example of costimulation blockers successfully exploited in renal transplantation. Due to the wide range of regulatory molecules characterized in the past decades, some of these molecules might be candidates as immunomodulators in the case of tolerance induction in transplantation. Although there are numerous attempts on the apprehension of the effects of co-signaling molecules on immune response, the necessity for a better understanding is evident. By increasing the knowledge on the biology of co-signaling pathways, some pitfalls are recognized and improved approaches are proposed. The blockage of CD80/CD28 axis is an instance of evolution toward more efficacy. It is now evident that anti-CD28 antibodies are more effective than CD80 blockers in animal models of transplantation. Other co-signaling axes such as PD-1/PD-L1, CD40/CD154, 2B4/CD48, and others discussed in the present review are examples of critical immunomodulatory molecules in allogeneic transplantation. We review here the outcomes of recent experiences with co-signaling molecules in preclinical studies of solid organ transplantation.
Collapse
Affiliation(s)
- Alireza Mardomi
- Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran.,Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nabiallah Mohammadi
- Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Saeid Abediankenari
- Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
17
|
Yu S, Su C, Luo X. Impact of infection on transplantation tolerance. Immunol Rev 2019; 292:243-263. [PMID: 31538351 PMCID: PMC6961566 DOI: 10.1111/imr.12803] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/29/2019] [Accepted: 09/03/2019] [Indexed: 12/12/2022]
Abstract
Allograft tolerance is the ultimate goal of organ transplantation. Current strategies for tolerance induction mainly focus on inhibiting alloreactive T cells while promoting regulatory immune cells. Pathogenic infections may have direct impact on both effector and regulatory cell populations, therefore can alter host susceptibility to transplantation tolerance induction as well as impair the quality and stability of tolerance once induced. In this review, we will discuss existing data demonstrating the effect of infections on transplantation tolerance, with particular emphasis on the role of the stage of infection (acute, chronic, or latent) and the stage of tolerance (induction or maintenance) in this infection-tolerance interaction. While the deleterious effect of acute infection on tolerance is mainly driven by proinflammatory cytokines induced shortly after the infection, chronic infection may generate exhausted T cells that could in fact facilitate transplantation tolerance. In addition to pathogenic infections, commensal intestinal microbiota also has numerous significant immunomodulatory effects that can shape the host alloimmunity following transplantation. A comprehensive understanding of these mechanisms is crucial for the development of therapeutic strategies for robustly inducing and stably maintaining transplantation tolerance while preserving host anti-pathogen immunity in clinically relevant scenarios.
Collapse
Affiliation(s)
- Shuangjin Yu
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, United States
- Division of Organ transplantation, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Chang Su
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, United States
| | - Xunrong Luo
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, United States
- Duke Transplant Center, Duke University School of Medicine, Durham, NC 27710, United States
| |
Collapse
|
18
|
Wong K, Shen J, D’Ambruoso S, Stefanoudakis D, Drakaki A. Safety and Efficacy of Immune Checkpoint Inhibitors in Patients With Metastatic Cancer Post Solid Organ Transplantation: A Case Report and Review of the Literature. Transplant Proc 2019; 51:3053-3058. [DOI: 10.1016/j.transproceed.2019.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/13/2019] [Accepted: 08/13/2019] [Indexed: 12/25/2022]
|
19
|
Hu B, Yang XB, Sang XT. Liver graft rejection following immune checkpoint inhibitors treatment: a review. Med Oncol 2019; 36:94. [DOI: 10.1007/s12032-019-1316-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023]
Abstract
Abstract
Immune checkpoint inhibitors (ICIs) have demonstrated remarkable efficacy in a variety of solid tumors; nonetheless, they have not been well investigated and are still recognized as a relative contraindication for patients with a liver transplantation (LT) history, since ICIs treatment might potentially lead to graft rejection. The program death-1 (PD-1) and the cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) pathways are implicated in the tolerance of transplanted organ, as well as blockade of the pathways, which contribute to eliminating tumors and may inadvertently lead to peripheral transplant rejection. Currently, no guidelines are available regarding the treatment for ICIs patients with a prior LT history. Therefore, this study was carried out to review the recent studies, attempting to introduce the ICIs-related graft rejection after LT from various aspects. We believed that ICIs could be given for the well-informed patients receiving LT and developed recurrence in a controlled setting. Typically, these patients should be treated according to a clinical care path or a prospective clinical trial, so as obtain a persistent anti-tumor immune response in the meantime of avoiding graft rejection, adjust the immunosuppression, reduce the possibility of graft loss following rejection, and have the opportunity to develop biomarkers for tumor response and transplant rejection.
Collapse
|
20
|
Selective Costimulation Blockade With Antagonist Anti-CD28 Therapeutics in Transplantation. Transplantation 2019; 103:1783-1789. [DOI: 10.1097/tp.0000000000002740] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
21
|
Crepeau R, Elengickal J, La Muraglia G, Ford M. Impact of selective CD28 blockade on virus-specific immunity to a murine Epstein-Barr virus homolog. Am J Transplant 2019; 19:2199-2209. [PMID: 30801917 PMCID: PMC6658342 DOI: 10.1111/ajt.15321] [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: 10/03/2018] [Revised: 01/13/2019] [Accepted: 01/31/2019] [Indexed: 01/25/2023]
Abstract
CTLA-4Ig (belatacept) blocks the CD80/CD86 ligands for both CD28 and CTLA-4; thus, in addition to the intended effect of blocking CD28-mediated costimulation, belatacept also has the unintended effect of blocking CTLA-4-mediated coinhibition. Recently, anti-CD28 domain antibodies (dAb) that selectively target CD28 while leaving CTLA-4 intact were shown to more effectively inhibit alloimmune responses and prolong graft survival. However, the impact of selective CD28 blockade on protective immunity has not been extensively investigated. Here, we sought to compare the impact of CTLA-4Ig vs anti-CD28dAb on CD8+ T cell immunity to a transplant-relevant pathogen, a murine homolog of Epstein-Barr virus. Mice were infected with murine gammaherpesvirus-68 (MHV) and treated with vehicle, CTLA-4Ig, or anti-CD28dAb. Although anti-CD28dAb resulted in a decrease in virus-specific CD8+ T cell numbers as compared to CTLA-4Ig, cytolytic function and the expression of markers of high-quality effectors were not different from CTLA-4Ig treated animals. Importantly, MHV-68 viral load was not different between the treatment groups. These results suggest that preserved CTLA-4 coinhibition limits MHV-specific CD8+ T cell accumulation, but the population that remains retains cytolytic function and migratory capacity and is not inferior in its ability to control viral burden relative to T cell responses in CTLA-4Ig-treated animals.
Collapse
Affiliation(s)
- R.L. Crepeau
- Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, Georgia
| | - J.A. Elengickal
- Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, Georgia
| | - G.M. La Muraglia
- Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, Georgia
| | - M.L. Ford
- Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
22
|
Czaja AJ. Immune inhibitory proteins and their pathogenic and therapeutic implications in autoimmunity and autoimmune hepatitis. Autoimmunity 2019; 52:144-160. [PMID: 31298041 DOI: 10.1080/08916934.2019.1641200] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Key inhibitory proteins can blunt immune responses to self-antigens, and deficiencies in this repertoire may promote autoimmunity. The goals of this review are to describe the key immune inhibitory proteins, indicate their possible impact on the development of autoimmune disease, especially autoimmune hepatitis, and encourage studies to clarify their pathogenic role and candidacy as therapeutic targets. English abstracts were identified in PubMed by multiple search terms. Full length articles were selected for review, and secondary and tertiary bibliographies were developed. Cytotoxic T lymphocyte antigen-4 impairs ligation of CD28 to B7 ligands on antigen presenting cells and inhibits the adaptive immune response by increasing anti-inflammatory cytokines, generating regulatory T cells, and reducing T cell activation and proliferation. Programed cell death antigen-1 inhibits T cell selection, activation, and proliferation by binding with two ligands at different phases and locations of the immune response. A soluble alternatively spliced variant of this protein can dampen the inhibitory signal. Autoimmune hepatitis has been associated with polymorphisms of the cytotoxic T lymphocyte antigen-4 gene, reduced hepatic expression of a ligand of programed cell death antigen-1, an interfering soluble variant of this key inhibitory protein, and antibodies against it. Findings have been associated with laboratory indices of liver injury and suboptimal treatment response. Abatacept, belatacept, CD28 blockade, and induction of T cell exhaustion are management considerations that require scrutiny. In conclusion, deficiencies in key immune inhibitory proteins may promote the occurrence of autoimmune diseases, such as autoimmune hepatitis, and emerging interventions may overcome these deficiencies. Investigations should define the nature, impact and management of these inhibitory disturbances in autoimmune hepatitis.
Collapse
Affiliation(s)
- Albert J Czaja
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| |
Collapse
|
23
|
Kennedy LB, Salama AKS. A Review of Immune-Mediated Adverse Events in Melanoma. Oncol Ther 2019; 7:101-120. [PMID: 32699983 PMCID: PMC7359990 DOI: 10.1007/s40487-019-0096-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Indexed: 12/16/2022] Open
Abstract
The use of checkpoint inhibitor-based immunotherapy has transformed the treatment landscape for melanoma as well as many other cancer types. With the ability to potentiate tumor-specific immune responses, these agents can result in durable tumor control. However, this activation of the immune system can lead to a unique constellation of side effects, distinct from other cancer therapies, collectively termed immune-mediated adverse events (irAEs). This review will focus on irAEs and guidelines for management related to the most clinically relevant checkpoint inhibitors, those that target programmed death receptor-1 (PD-1) and cytotoxic T lymphocyte antigen-4 (CTLA-4).
Collapse
|
24
|
Kaufman CL, Bhutiani N, Ramirez A, Tien HY, Palazzo MD, Galvis E, Farner S, Ozyurekoglu T, Jones CM. Current Status of Vascularized Composite Allotransplantation. Am Surg 2019. [DOI: 10.1177/000313481908500628] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The field of vascularized composite allotransplantation (VCA) has moved from a highly experimental procedure to, at least for some patients, one of the best treatment alternatives for catastrophic tissue loss or dysfunction. Although the worldwide experience is still limited, progress has been made in translation to the clinic, and hand transplantation was recently designated standard of care and is now covered in full by the British Health System. This progress is tempered by the long-term challenges of systemic immunosuppression, and the rapidly evolving indications for VCA such as urogenital transplantation. This update will cover the state of and recent changes in the field, and an update of the Louisville VCA program as our initial recipient, the first person to receive a hand transplant in the United States celebrates the 20th anniversary of his transplant. The achievements and complications encountered over the last two decades will be reviewed. In addition, potential directions for research and collaboration as well as practical issues of how third party payers and funding are affecting growth of the field are presented.
Collapse
|
25
|
Abstract
Immune checkpoint inhibitors (ICIs) are therapeutic antibodies that target regulatory molecules on T cells and represent the most widely used FDA-approved class of immunotherapy. ICIs are associated with unique immune-mediated toxicities called immune-related adverse events. These toxicities may affect any organ system, and their precise mechanisms of action remain under investigation. Current evidence suggests that activation of T cells is involved, although other components of the immune response have been implicated. This article summarizes toxicities, potential mechanisms of action, management strategies, and other clinical considerations. Unique mechanisms of action and immune-related toxicities of other FDA-approved classes of immunotherapy are reviewed.
Collapse
Affiliation(s)
- Katherine Sanchez
- Earle A. Chiles Research Institute, 4805 Northeast Glisan Street, North Pavilion, 2N, Portland, OR 97213, USA.
| | - David B Page
- Earle A. Chiles Research Institute, 4805 Northeast Glisan Street, North Pavilion, 2N, Portland, OR 97213, USA
| | - Walter Urba
- Earle A. Chiles Research Institute, 4805 Northeast Glisan Street, North Pavilion, 2N, Portland, OR 97213, USA
| |
Collapse
|
26
|
Au KP, Chok KSH. Multidisciplinary approach for post-liver transplant recurrence of hepatocellular carcinoma: A proposed management algorithm. World J Gastroenterol 2018; 24:5081-5094. [PMID: 30568386 PMCID: PMC6288653 DOI: 10.3748/wjg.v24.i45.5081] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 10/21/2018] [Accepted: 11/07/2018] [Indexed: 02/06/2023] Open
Abstract
A large number of liver transplants have been performed for hepatocellular carcinoma (HCC), and recurrence is increasingly encountered. The recurrence of HCC after liver transplantation is notoriously difficult to manage. We hereby propose multi-disciplinary management with a systematic approach. The patient is jointly managed by the transplant surgeon, physician, oncologist and radiologist. Immunosuppressants should be tapered to the lowest effective dose to protect against rejection. The combination of a mammalian target of rapamycin inhibitor with a reduced calcineurin inhibitor could be considered with close monitoring of graft function and toxicity. Comprehensive staging can be performed by dual-tracer positron emission tomography-computed tomography or the combination of contrast computed tomography and a bone scan. In patients with disseminated recurrence, sorafenib confers survival benefits but is associated with significant drug toxicity. Oligo-recurrence encompasses recurrent disease that is limited in number and location so that loco-regional treatments convey disease control and survival benefits. Intra-hepatic recurrence can be managed with graft resection, but significant operative morbidity is expected. Radiofrequency ablation and stereotactic body radiation therapy (SBRT) are effective alternative strategies. In patients with more advanced hepatic disease, regional treatment with trans-arterial chemoembolization or intra-arterial Yttrium-90 can be considered. For patients with extra-hepatic oligo-recurrence, loco-regional treatment can be considered if practical. Patients with more than one site of recurrence are not always contraindicated for curative treatments. Surgical resection is effective for patients with pulmonary oligo-recurrence, but adequate lung function is a pre-requisite. SBRT is a non-invasive and effective modality that conveys local control to pulmonary and skeletal oligo-recurrences.
Collapse
Affiliation(s)
- Kin Pan Au
- Department of Surgery, Queen Mary Hospital, Hong Kong, China
| | - Kenneth Siu Ho Chok
- Department of Surgery and State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
27
|
Selective CD28 Inhibition Modulates Alloimmunity and Cardiac Allograft Vasculopathy in Anti-CD154-Treated Monkeys. Transplantation 2018; 102:e90-e100. [PMID: 29319621 DOI: 10.1097/tp.0000000000002044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Selective CD28 inhibition is actively pursued as an alternative to B7 blockade using cytotoxic T lymphocyte antigen 4 Ig based on the hypothesis that the checkpoint immune regulators cytotoxic T lymphocyte antigen 4 and programmed death ligand 1 will induce tolerogenic immune signals. We previously showed that blocking CD28 using a monovalent nonactivating reagent (single-chain anti-CD28 Fv fragment linked to alpha-1 antitrypsin [sc28AT]) synergizes with calcineurin inhibitors in nonhuman primate (NHP) kidney and heart transplantation. Here, we explored the efficacy of combining a 3-week "induction" sc28AT treatment with prolonged CD154 blockade. METHODS Cynomolgus monkey heterotopic cardiac allograft recipients received sc28AT (10 mg/kg, d0-20, n = 3), hu5C8 (10-30 mg/kg, d0-84, n = 4), or combination (n = 6). Graft survival was monitored by telemetry. Protocol biopsies and graft explants were analyzed for International Society of Heart and Lung Transplantation acute rejection grade and cardiac allograft vasculopathy score. Alloantibody, T-cell phenotype and regulatory T cells were analyzed by flow cytometry. Immunochemistry and gene expression (NanoString) characterized intra-graft cellular infiltration. RESULTS Relative to modest prolongation of median graft survival time with sc28AT alone (34 days), hu5C8 (133 days), and sc28AT + hu5C8 (141 days) prolonged survival to a similar extent. CD28 blockade at induction, added to hu5C8, significantly attenuated the severity of acute rejection and cardiac allograft vasculopathy during the first 3 months after transplantation relative to hu5C8 alone. These findings were associated with decreased proportions of circulating CD8 and CD3CD28 T cells, and modulation of inflammatory gene expression within allografts. CONCLUSIONS Induction with sc28AT promotes early cardiac allograft protection in hu5C8-treated NHPs. These results support further investigation of prolonged selective CD28 inhibition with CD40/CD154 blockade in NHP transplants.
Collapse
|
28
|
Grant MJ, DeVito N, Salama AKS. Checkpoint inhibitor use in two heart transplant patients with metastatic melanoma and review of high-risk populations. Melanoma Manag 2018; 5:MMT10. [PMID: 30459942 PMCID: PMC6240846 DOI: 10.2217/mmt-2018-0004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 08/03/2018] [Indexed: 12/12/2022] Open
Abstract
Due to the unique side-effect profile of immune checkpoint inhibitors (ICIs), groups of patients deemed to be at high risk of complications were excluded from trials that proved the efficacy and safety of these agents in patients with various malignancies. Among these excluded patients were those with prior solid organ transplantation, chronic viral infections and pre-existing autoimmune diseases including paraneoplastic syndromes. We present follow-up on a patient from a previously published case report with an orthotopic heart transplantation who was treated with both cytotoxic T-lymphocyte antigen 4 and PD-1 inhibition safely, without organ rejection. Additionally, we describe the case of a patient with a cardiac allograft who also did not experience organ rejection after treatment with pembrolizumab. Through smaller trials, retrospective analyses, case series and individual case reports, we are accumulating initial data on how these agents are tolerated by the aforementioned groups. Our survey of the literature has found more evidence of organ transplant rejection in patients treated with PD-1 inhibitors than those treated with inhibitors of cytotoxic T-lymphocyte antigen 4. Patients with chronic viral infections, especially hepatitis C, seem to have little to no risk of treatment-related increase in serum RNA levels. The literature contains few documented cases of devastating exacerbations of pre-existing autoimmune disease during treatment with ICIs, and flares seem to be easily controlled by immunosuppression in the vast majority of cases. Last, several cases allude to a promising role for disease-specific antibodies and other serum biomarkers in identifying patients at high risk of developing certain immune-related adverse events, detecting subclinical immune-related adverse event onset, and monitoring treatment response to immunosuppressive therapy in patients treated with ICIs. Though these excluded populations have not been well studied in randomized placebo-controlled trials, we may be able to learn and derive hypotheses from the existing observational data in the literature.
Collapse
Affiliation(s)
- Michael J Grant
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.,Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Nicholas DeVito
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA.,Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - April K S Salama
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA.,Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
29
|
Costello R, Kissenpfennig A, Martins PN, McDaid J. Development of transplant immunosuppressive agents - considerations in the use of animal models. Expert Opin Drug Discov 2018; 13:1041-1053. [PMID: 30332905 DOI: 10.1080/17460441.2018.1535589] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
INTRODUCTION The development of all immunosuppressant agents to date has involved the experimental use of large and small animal models. Over the last half-century, immunosuppressive drugs have extended the lives of transplant patients worldwide. However, the use of animal models in the development of these drugs is not perfect, and this has brought to light a number of issues including idiosyncratic reactions that are found in animal models but not in humans. The 2006 highly publicized case of the 'elephant man' TGN 1412 drug trial highlights the importance of being cogent of the limitations of animal models. Areas covered: This review covers the utility and limitations of the use of animal models for the development of immunosuppressant agents. This includes both large and small animal models, particularly rodent models in the transplant setting. Expert opinion: The use of animal models represents a critical stage in the development of immunosuppressive drugs. Limitations include physiological differences to humans; this is especially true of immunologically naïve lab rodents with small memory cell populations. Toxic drug levels may differ widely between species. Animal models are also costly and raise ethical concerns. However, there is currently no way to recreate the complex environment of the human immune system purely in vitro.
Collapse
Affiliation(s)
- Russell Costello
- a Wellcome Wolfson Institute for Experimental Medicine , Queen's University , Belfast , UK
| | - Adrien Kissenpfennig
- a Wellcome Wolfson Institute for Experimental Medicine , Queen's University , Belfast , UK
| | - Paulo N Martins
- b Department of Surgery, Division of Transplantation, UMass Memorial Medical Center , University of Massachusetts , Worchester , MA , USA
| | - James McDaid
- c Department of Transplant Surgery , City Hospital , Belfast , UK
| |
Collapse
|
30
|
Babey H, Quéré G, Descourt R, Le Calloch R, Lanfranco L, Nousbaum JB, Cornec D, Tison A, Chouaid C. Immune-checkpoint inhibitors to treat cancers in specific immunocompromised populations: a critical review. Expert Rev Anticancer Ther 2018; 18:981-989. [PMID: 29995451 DOI: 10.1080/14737140.2018.1499468] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Because of their efficacy against numerous cancers, immune-checkpoint inhibitors (ICIs), anti-cytotoxic T-lymphocyte antigen-4, and anti-programmed cell death monoclonal antibodies are being used ever more often in oncology. However, some patients were excluded from clinical trials because of their comorbidities despite their potentially higher cancer frequencies, as is the case for immunocompromised patients. Areas covered: We analyzed reported preclinical and clinical information and evaluated the risk/benefit ratio for four immunocompromised populations: people living with human immunodeficiency virus (PLHs), solid-organ transplant recipients, recipients of hematopoietic stem-cell allografts, and patients with autoimmune diseases. Expert commentary: Information available in the literature is fragmentary and scarce, making it difficult to evaluate the risk/benefit ratio. It can, nonetheless, be noted that ICI use in PLHs seems possible. For solid-organ transplant recipients, the risk for the graft seems elevated. For the other two populations, it is difficult to conclude at this time.
Collapse
Affiliation(s)
- Hélène Babey
- a Institut de Cancerologie de Bretagne occidentale , Centre Hospitalier Universitaire de Brest , Brest , France
| | - Gilles Quéré
- a Institut de Cancerologie de Bretagne occidentale , Centre Hospitalier Universitaire de Brest , Brest , France
| | - Renaud Descourt
- a Institut de Cancerologie de Bretagne occidentale , Centre Hospitalier Universitaire de Brest , Brest , France
| | - Ronan Le Calloch
- b Service des maladies du sang, médecine interne, maladies infectieuses (MIIS) , Centre Hospitalier de Quimper Cornouaille , Quimper , France.,c Université de Brest , Fédération Inter Hospitalier d'Immuno-Hématologie de Bretagne Occidentale (FIHBO) , Brest , France
| | - Luca Lanfranco
- d Service de néphrologie , Centre Hospitalier Universitaire de Brest , Brest , France.,e UMR1227, Lymphocytes B et Autoimmunité, Inserm, LabEx IGO , Université de Brest , Brest , France
| | - Jean-Baptiste Nousbaum
- f Service d'Hépato-gastroentérologie , Centre Hospitalier Universitaire de Brest , Brest , France.,g Registre Finistérien des Tumeurs Digestives, EA 7479 SPURBO , Université de Bretagne Occidentale , Brest , France
| | - Divi Cornec
- e UMR1227, Lymphocytes B et Autoimmunité, Inserm, LabEx IGO , Université de Brest , Brest , France.,h Rhumatologie et Centre National de Référence des Maladies Auto-Immunes Rares CERAINO , CHRU de Brest , Brest , France
| | - Alice Tison
- e UMR1227, Lymphocytes B et Autoimmunité, Inserm, LabEx IGO , Université de Brest , Brest , France.,h Rhumatologie et Centre National de Référence des Maladies Auto-Immunes Rares CERAINO , CHRU de Brest , Brest , France
| | - Christos Chouaid
- i Service de pneumologie , Centre Hospitalier Intercommunal de Créteil , Créteil , France
| |
Collapse
|
31
|
Schwarz C, Mahr B, Muckenhuber M, Wekerle T. Belatacept/CTLA4Ig: an update and critical appraisal of preclinical and clinical results. Expert Rev Clin Immunol 2018; 14:583-592. [PMID: 29874474 DOI: 10.1080/1744666x.2018.1485489] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The B7/CD28/CTLA4 signaling cascade is the most thoroughly studied costimulatory pathway and blockade with CTLA4Ig (abatacept) or its derivative belatacept has emerged as a valuable option for pharmacologic immune modulation. Several clinical studies have ultimately led to the approval of belatacept for immunosuppression in kidney transplant recipients. Areas covered: This review will discuss the immunological background of costimulation blockade and recent preclinical data and clinical results of CTLA4Ig/belatacept. Expert commentary: The development of belatacept is a major advance in clinical transplantation. However, in spite of promising results in preclinical and clinical trials, clinical use remains limited at present, in part due to increased rates of acute rejection. Recent efforts showing encouraging progress in refining such protocols might be a step toward harnessing the full potential of costimulation blockade-based immunosuppression.
Collapse
Affiliation(s)
- Christoph Schwarz
- a Division of General Surgery, Department of Surgery , Medical University of Vienna , Vienna , Austria.,b Section of Transplantation Immunology, Department of Surgery , Medical University of Vienna , Vienna , Austria
| | - Benedikt Mahr
- b Section of Transplantation Immunology, Department of Surgery , Medical University of Vienna , Vienna , Austria
| | - Moritz Muckenhuber
- b Section of Transplantation Immunology, Department of Surgery , Medical University of Vienna , Vienna , Austria
| | - Thomas Wekerle
- b Section of Transplantation Immunology, Department of Surgery , Medical University of Vienna , Vienna , Austria
| |
Collapse
|
32
|
Abstract
Modulation of T-cell activity through blockade of coinhibitory molecules has revolutionized the treatment of various malignancies. Several immune checkpoint inhibitors are currently Food and Drug Administration approved which target various coinhibitory pathways including cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed death 1 receptor (PD-1), and programmed cell death ligand-1. Clinical trials that lead to the Food and Drug Administration approval of these agents often excluded patients with an organ transplant. Excluding these patients was deliberate due to concern that immune checkpoint inhibitor therapy could lead to graft rejection. The PD-1 and CTLA-4 pathways are essential to downregulate our immune system in the setting of T-cell activation to prevent autoimmunity. Furthermore, both pathways are implicated in transplanted organ tolerance and modulation of the pathways may inadvertently lead to peripheral transplant rejection. Currently, there are no guidelines for the treatment of patients with immune checkpoint inhibitors in the setting of a prior organ transplant. Thus far, there are only 10 reported cases of patients in the literature who were treated in this setting. Two additional cases are reported herein, including 1 patient with a prior cardiac transplant receiving nivolumab for non-small cell lung cancer. Of the 12 cases, 4 patients experienced organ rejection. From these observations, the authors hypothesize factors that affect safety and of this treatment modality in this patient population. These factors include the integral role of the PD-1 pathway compared with the CTLA-4 pathway in organ acceptance, sequential implementation of different immune checkpoint inhibitor classes, length of time with a transplant before therapy, strength of immunosuppressive agents to prevent organ transplant rejection, and immunogenicity of the particular organ grafted. Although limited cases have been reported, there are circumstances in which immune checkpoint inhibitors have been used in the setting of organ transplantation without resulting in organ rejection. A thorough discussion with the patient of the potential risks, including graft rejection, and benefits of this therapy is necessary before beginning this treatment. More research is needed to explore the safety and efficacy of immune checkpoint inhibitors in the setting of organ transplantation.
Collapse
|
33
|
Singh SS, Jois SD. Homo- and Heterodimerization of Proteins in Cell Signaling: Inhibition and Drug Design. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2018; 111:1-59. [PMID: 29459028 DOI: 10.1016/bs.apcsb.2017.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Protein dimerization controls many physiological processes in the body. Proteins form homo-, hetero-, or oligomerization in the cellular environment to regulate the cellular processes. Any deregulation of these processes may result in a disease state. Protein-protein interactions (PPIs) can be inhibited by antibodies, small molecules, or peptides, and inhibition of PPI has therapeutic value. PPI drug discovery research has steadily increased in the last decade, and a few PPI inhibitors have already reached the pharmaceutical market. Several PPI inhibitors are in clinical trials. With advancements in structural and molecular biology methods, several methods are now available to study protein homo- and heterodimerization and their inhibition by drug-like molecules. Recently developed methods to study PPI such as proximity ligation assay and enzyme-fragment complementation assay that detect the PPI in the cellular environment are described with examples. At present, the methods used to design PPI inhibitors can be classified into three major groups: (1) structure-based drug design, (2) high-throughput screening, and (3) fragment-based drug design. In this chapter, we have described some of the experimental methods to study PPIs and their inhibition. Examples of homo- and heterodimers of proteins, their structural and functional aspects, and some of the inhibitors that have clinical importance are discussed. The design of PPI inhibitors of epidermal growth factor receptor heterodimers and CD2-CD58 is discussed in detail.
Collapse
Affiliation(s)
- Sitanshu S Singh
- Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA, United States
| | - Seetharama D Jois
- Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA, United States.
| |
Collapse
|
34
|
Liu D, Badell IR, Ford ML. Selective CD28 blockade attenuates CTLA-4-dependent CD8+ memory T cell effector function and prolongs graft survival. JCI Insight 2018; 3:96378. [PMID: 29321374 DOI: 10.1172/jci.insight.96378] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 11/28/2017] [Indexed: 12/21/2022] Open
Abstract
Memory T cells pose a significant problem to successful therapeutic control of unwanted immune responses during autoimmunity and transplantation, as they are differentially controlled by cosignaling receptors such as CD28 and CTLA-4. Treatment with abatacept and belatacept impede CD28 signaling by binding to CD80 and CD86, but they also have the unintended consequence of blocking the ligands for CTLA-4, a process that may inadvertently boost effector responses. Here, we show that a potentially novel anti-CD28 domain antibody (dAb) that selectively blocks CD28 but preserves CTLA-4 coinhibition confers improved allograft survival in sensitized recipients as compared with CTLA-4 Ig. However, both CTLA-4 Ig and anti-CD28 dAb similarly and significantly reduced the accumulation of donor-reactive CD8+ memory T cells, demonstrating that regulation of the expansion of CD8+ memory T cell populations is controlled in part by CD28 signals and is not significantly impacted by CTLA-4. In contrast, selective CD28 blockade was superior to CTLA-4 Ig in inhibiting IFN-γ, TNF, and IL-2 production by CD8+ memory T cells, which in turn resulted in reduced recruitment of innate CD11b+ monocytes into allografts. Importantly, this superiority was CTLA-4 dependent, demonstrating that effector function of CD8+ memory T cells is regulated by the balance of CD28 and CTLA-4 signaling.
Collapse
|
35
|
Vanhove B, Poirier N, Fakhouri F, Laurent L, 't Hart B, Papotto PH, Rizzo LV, Zaitsu M, Issa F, Wood K, Soulillou JP, Blancho G. Antagonist Anti-CD28 Therapeutics for the Treatment of Autoimmune Disorders. Antibodies (Basel) 2017; 6:antib6040019. [PMID: 31548534 PMCID: PMC6698823 DOI: 10.3390/antib6040019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 11/16/2017] [Accepted: 11/18/2017] [Indexed: 12/17/2022] Open
Abstract
The effector functions of T lymphocytes are responsible for most autoimmune disorders and act by directly damaging tissues or by indirectly promoting inflammation and antibody responses. Co-stimulatory and co-inhibitory T cell receptor molecules are the primary pharmacological targets that enable interference with immune-mediated diseases. Among these, selective CD28 antagonists have drawn special interest, since they tip the co-stimulation/co-inhibition balance towards efficiently inhibiting effector T cells while promoting suppression by pre-existing regulatory T-cells. After having demonstrated outstanding therapeutic efficacy in multiple models of autoimmunity, inflammation and transplantation, and safety in phase-I studies in humans, selective CD28 antagonists are currently in early clinical development for the treatment of systemic lupus erythematous and rheumatoid arthritis. Here, we review the available proof of concept studies for CD28 antagonists in autoimmunity, with a special focus on the mechanisms of action.
Collapse
Affiliation(s)
- Bernard Vanhove
- OSE Immunotherapeutics, 44200 Nantes, France.
- Centre de Recherche en Transplantation et Immunologie (CRTI) UMR1064, INSERM, Université de Nantes, 44035 Nantes, France.
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, 44093 Nantes, France.
| | - Nicolas Poirier
- OSE Immunotherapeutics, 44200 Nantes, France.
- Centre de Recherche en Transplantation et Immunologie (CRTI) UMR1064, INSERM, Université de Nantes, 44035 Nantes, France.
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, 44093 Nantes, France.
| | - Fadi Fakhouri
- Centre de Recherche en Transplantation et Immunologie (CRTI) UMR1064, INSERM, Université de Nantes, 44035 Nantes, France.
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, 44093 Nantes, France.
| | - Laetitia Laurent
- Centre de Recherche en Transplantation et Immunologie (CRTI) UMR1064, INSERM, Université de Nantes, 44035 Nantes, France.
| | - Bert 't Hart
- Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands.
- Department Neuroscience, University of Groningen, University Medical Center, 9713 GZ Groningen, The Netherlands.
| | - Pedro H Papotto
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal.
| | - Luiz V Rizzo
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627-701, 2-SS Bloco A, 05651-901 São Paulo, Brazil.
| | - Masaaki Zaitsu
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK.
| | - Fadi Issa
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK.
| | - Kathryn Wood
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK.
| | - Jean-Paul Soulillou
- Centre de Recherche en Transplantation et Immunologie (CRTI) UMR1064, INSERM, Université de Nantes, 44035 Nantes, France.
| | - Gilles Blancho
- Centre de Recherche en Transplantation et Immunologie (CRTI) UMR1064, INSERM, Université de Nantes, 44035 Nantes, France.
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, 44093 Nantes, France.
| |
Collapse
|
36
|
Zaitsu M, Issa F, Hester J, Vanhove B, Wood KJ. Selective blockade of CD28 on human T cells facilitates regulation of alloimmune responses. JCI Insight 2017; 2:89381. [PMID: 28978798 DOI: 10.1172/jci.insight.89381] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 08/29/2017] [Indexed: 01/05/2023] Open
Abstract
T cells are central to the detrimental alloresponses that develop in autoimmunity and transplantation, with CD28 costimulatory signals being key to T cell activation and proliferation. CTLA4-Ig molecules that bind CD80/86 and inhibit CD28 costimulation offer an alternative immunosuppressive treatment, free from some of the chronic toxicities associated with calcineurin inhibition. However, CD80/86 blockade by CTLA4-Ig also results in the loss of coinhibitory CTLA4 signals that are critical to the regulation of T cell activation. Here, we show that a nonactivating monovalent anti-CD28 that spares CTLA4 signaling is an effective immunosuppressant in a clinically relevant humanized mouse transplant model. We demonstrate that selective CD28 blockade prolongs human skin allograft survival through a mechanism that includes a reduction in the cellular graft infiltrate. Critically, selective CD28 blockade promotes Treg function in vivo and synergizes with adoptive Treg therapy to promote transplant survival. In contrast to CTLA4-Ig treatment, selective CD28 blockade promotes regulation of alloimmune responses and facilitates Treg-based cellular therapy.
Collapse
Affiliation(s)
- Masaaki Zaitsu
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom.,Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Fadi Issa
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Joanna Hester
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Bernard Vanhove
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,OSE Immunotherapeutics, Nantes, France
| | - Kathryn J Wood
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| |
Collapse
|
37
|
Dillinger B, Ahmadi-Erber S, Soukup K, Halfmann A, Schrom S, Vanhove B, Steinberger P, Geyeregger R, Ladisch S, Dohnal AM. CD28 Blockade Ex Vivo Induces Alloantigen-Specific Immune Tolerance but Preserves T-Cell Pathogen Reactivity. Front Immunol 2017; 8:1152. [PMID: 28979262 PMCID: PMC5611377 DOI: 10.3389/fimmu.2017.01152] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/31/2017] [Indexed: 12/21/2022] Open
Abstract
Donor T-cells contribute to reconstitution of protective immunity after allogeneic hematopoietic stem cell transplantation (HSCT) but must acquire specific tolerance against recipient alloantigens to avoid life-threatening graft-versus-host disease (GvHD). Systemic immunosuppressive drugs may abrogate severe GvHD, but this also impedes memory responses to invading pathogens. Here, we tested whether ex vivo blockade of CD28 co-stimulation can enable selective T-cell tolerization to alloantigens by facilitating CD80/86-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) signaling. Treatment of human allogeneic dendritic cell/T-cell co-cultures with a human CD28 blocking antibody fragment (α-huCD28) significantly abrogated subsequent allospecific immune responses, seen by decreased T-cell proliferation and of type 1 cytokine (IFN-γ and IL-2) expression. Allo-tolerization persisted after discontinuation of CD28 blockade and secondary alloantigen stimulation, as confirmed by enhanced CTLA-4 and PD-1 immune checkpoint signaling. However, T-cells retained reactivity to pathogens, supported by clonotyping of neo-primed and cross-reactive T-cells specific for Candida albicans or third-party antigens using deep sequencing analysis. In an MHC-mismatched murine model, we tolerized C57BL/6 T-cells by ex vivo exposure to a murine single chain Fv specific for CD28 (α-muCD28). Infusion of these cells, after α-muCD28 washout, into bone marrow-transplanted BALB/c mice caused allo-tolerance and did not induce GvHD-associated hepatic pathology. We conclude that selective CD28 blockade ex vivo can allow the generation of stably allo-tolerized T-cells that in turn do not induce graft-versus-host reactions while maintaining pathogen reactivity. Hence, CD28 co-stimulation blockade of donor T-cells may be a useful therapeutic approach to support the immune system after HSCT.
Collapse
Affiliation(s)
- Barbara Dillinger
- Tumor Immunology, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung e.V., Vienna, Austria
| | - Sarah Ahmadi-Erber
- Tumor Immunology, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung e.V., Vienna, Austria
| | - Klara Soukup
- Tumor Immunology, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung e.V., Vienna, Austria
| | - Angela Halfmann
- Tumor Immunology, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung e.V., Vienna, Austria
| | - Silke Schrom
- Tumor Immunology, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung e.V., Vienna, Austria
| | - Bernard Vanhove
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,OSE Immunotherapeutics, Nantes, France
| | - Peter Steinberger
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Rene Geyeregger
- Clinical Cell Biology, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung e.V., Vienna, Austria.,Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Stephan Ladisch
- Center for Cancer and Immunology Research, Children's Research Institute, Children's National Medical Center, Washington, DC, United States
| | - Alexander Michael Dohnal
- Tumor Immunology, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung e.V., Vienna, Austria
| |
Collapse
|
38
|
Laurent L, Le Fur A, Le Bloas R, Néel M, Mary C, Moreau A, Poirier N, Vanhove B, Fakhouri F. Prevention of lupus nephritis development in NZB/NZW mice by selective blockade of CD28. Eur J Immunol 2017. [DOI: 10.1002/eji.201746923] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Laetitia Laurent
- INSERM UMR 1064; Nantes France
- Institut de Transplantation Urologie Néphrologie (ITUN); Université de Nantes; Nantes France
| | - Awena Le Fur
- INSERM UMR 1064; Nantes France
- Institut de Transplantation Urologie Néphrologie (ITUN); Université de Nantes; Nantes France
- Department of nephrology and immunology; Centre Hospitalier Universitaire; Nantes France
| | - Rozenn Le Bloas
- INSERM UMR 1064; Nantes France
- Institut de Transplantation Urologie Néphrologie (ITUN); Université de Nantes; Nantes France
| | - Mélanie Néel
- INSERM UMR 1064; Nantes France
- Institut de Transplantation Urologie Néphrologie (ITUN); Université de Nantes; Nantes France
| | - Caroline Mary
- INSERM UMR 1064; Nantes France
- Institut de Transplantation Urologie Néphrologie (ITUN); Université de Nantes; Nantes France
- OSE Immunotherapeutics; Nantes France
| | - Anne Moreau
- Department of pathology; Centre Hospitalier Universitaire; Nantes France
| | - Nicolas Poirier
- INSERM UMR 1064; Nantes France
- Institut de Transplantation Urologie Néphrologie (ITUN); Université de Nantes; Nantes France
- OSE Immunotherapeutics; Nantes France
| | - Bernard Vanhove
- INSERM UMR 1064; Nantes France
- Institut de Transplantation Urologie Néphrologie (ITUN); Université de Nantes; Nantes France
- OSE Immunotherapeutics; Nantes France
| | - Fadi Fakhouri
- INSERM UMR 1064; Nantes France
- Institut de Transplantation Urologie Néphrologie (ITUN); Université de Nantes; Nantes France
- Department of nephrology and immunology; Centre Hospitalier Universitaire; Nantes France
| |
Collapse
|
39
|
Crepeau RL, Ford ML. Challenges and opportunities in targeting the CD28/CTLA-4 pathway in transplantation and autoimmunity. Expert Opin Biol Ther 2017; 17:1001-1012. [PMID: 28525959 DOI: 10.1080/14712598.2017.1333595] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION T cell activation is a complex process that requires multiple cell signaling pathways, including a primary recognition signal and additional costimulatory signals. One of the best-characterized costimulatory pathways includes the Ig superfamily members CD28 and CTLA-4 and their ligands CD80 and CD86. Areas covered: This review discusses past, current and future biological therapies that have been utilized to block the CD28/CTLA-4 cosignaling pathway in the settings of autoimmunity and transplantation, as well the challenges facing successful implementation of these therapies. Expert opinion: The development of CD28 blockers Abatacept and Belatacept provided a more targeted therapy approach for transplant rejection and autoimmune disease relative to calcineurin inhibitors and anti-proliferatives, but overall efficacy may be limited due to their collateral effect of simultaneously blocking CTLA-4 coinhibitory signals. As such, current investigations into the potential of selective CD28 blockade to block the costimulatory potential of CD28 while exploiting the coinhibitory effects of CTLA-4 are promising. However, as selective CD28 blockade inhibits the activity of both effector and regulatory T cells, an important goal for the future is the design of therapies that will maximize the attenuation of effector responses while preserving the suppressive function of T regulatory cells.
Collapse
Affiliation(s)
- Rebecca L Crepeau
- a Emory Transplant Center and Department of Surgery , Emory University , Atlanta , GA , USA
| | - Mandy L Ford
- a Emory Transplant Center and Department of Surgery , Emory University , Atlanta , GA , USA
| |
Collapse
|
40
|
Abstract
BACKGROUND MicroRNAs (miRNAs) are small noncoding RNA molecules that regulate the posttranscriptional expression of target genes and are important regulators in immune responses. Previous studies demonstrated that the miRNA, miR-182 was significantly increased during allograft rejection. Further, the transcription factor Forkhead box (FOX) protein 1, (FOXO1) was shown to be a target of miR-182. The aim of this study is to further examine the role of miR-182 in alloimmune responses. METHODS Transplantation of BALB/c cardiac allografts was performed in C57BL/6, miR-182, B6.129S-H2 (MHC II and CD4 T cell-deficient) and B6.129S2-Tap1 (MHC I and CD8 T cell-deficient) mice, with or without CTLA-4Ig administration. T cell phenotype, FOXO1 protein levels and graft infiltrating lymphocytes were determined in C57BL/6 or miR-182 mice by flow cytometric analysis, Western blot, and immunohistochemistry, respectively. RESULTS We now show that T cells, mainly CD4 are the main cellular source of miR-182 during allograft rejection. In the absence of miR-182, CTLA-4Ig treatment significantly increased allograft survival (31.5 days C57BL/6 vs 60 days miR-182; P < 0.01). Further, CTLA4-Ig treatment inhibits miR-182 expression, increases FOXO1 levels, and reduces the percentage of CD4CD44 T cells after transplantation. Fewer T cells infiltrate the cardiac allografts, and memory T cells are significantly decreased in allograft recipients deficient in miR-182 with CTLA4-Ig treatment (P < 0.01). CONCLUSIONS Our findings suggest that miR-182 contributes to the T-cell responses to alloantigen especially under costimulation blockade. Therapeutics that target specific miRNAs may prove beneficial in transplantation.
Collapse
|
41
|
Jose A, Yiannoullou P, Bhutani S, Denley H, Morton M, Picton M, Summers A, van Dellen D, Augustine T. Renal Allograft Failure After Ipilimumab Therapy for Metastatic Melanoma: A Case Report and Review of the Literature. Transplant Proc 2017; 48:3137-3141. [PMID: 27932166 DOI: 10.1016/j.transproceed.2016.07.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 07/26/2016] [Indexed: 02/07/2023]
Abstract
Transplant recipients are at an increased risk of malignant melanoma, a result of chronic immunosuppression. Ipilimumab is a newer biological agent targeting T lymphocytes to potentiate an immune response against melanoma, and the use of this agent results in a new adverse effect profile that the clinician must be aware of while a patient is on therapy. We report the case of a male renal transplant recipient who developed graft failure while treated with ipilimumab and minimal immunosuppressive therapy for metastatic ocular melanoma, with biopsy evidence of glomerulonephritis and acute rejection. We highlight the immunological side effects that can manifest from ipilimumab therapy and conclude that it did influence graft function in this patient. Our case illustrates the importance of weighing the risks and benefits to graft function and long-term survival as well as the importance of considering other treatment modalities in this specific group of melanoma patients.
Collapse
Affiliation(s)
- A Jose
- Department of Transplant Surgery, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom; University of Manchester, Manchester, United Kingdom.
| | - P Yiannoullou
- Department of Transplant Surgery, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - S Bhutani
- Department of Nephrology, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - H Denley
- Department of Histopathology, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - M Morton
- Department of Nephrology, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - M Picton
- Department of Nephrology, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - A Summers
- Department of Transplant Surgery, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - D van Dellen
- Department of Transplant Surgery, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom; University of Manchester, Manchester, United Kingdom
| | - T Augustine
- Department of Transplant Surgery, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom; University of Manchester, Manchester, United Kingdom
| |
Collapse
|
42
|
Jin J, Gong J, Lin B, Li Y, He Q. FcγRIIb expression on B cells is associated with treatment efficacy for acute rejection after kidney transplantation. Mol Immunol 2017; 85:283-292. [PMID: 28360016 DOI: 10.1016/j.molimm.2017.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 03/03/2017] [Accepted: 03/06/2017] [Indexed: 11/26/2022]
Abstract
BACKGROUND Fcγ receptors (FcγR) play a role in the acute rejection (AR) of organ transplants. FcγRIIB is an inhibitory FcγR expressed on B cells. Intravenous IgG (IVIG) and CD28 monoclonal antibody (mAb) have been shown to have immunomodulatory properties against AR. AIM To examine the association between FcγRIIB expression on B cell subpopulations and AR treatment efficacy. METHODS Male F344 rats were used as kidney donors and Lewis rats as recipients to establish models of renal transplantation. Rats were divided into five groups: sham, AR-PBS, AR-IVIG, AR-PNGase F-IVIG, and AR-CD28. Serum creatinine (Scr), blood urea nitrogen (BUN), and urine protein content were determined. Inflammatory markers were measured by ELISA, FcγR by western blotting, and spleen B cell activation by flow cytometry. RESULTS Scr, BUN, urinary protein content, levels of CRP, IL-10, TNF-α, IL-6, IL-8, and IgG were all increased in the AR-PBS group compared with the sham group (all P<0.01); these increases were partly reversed in the AR-IVIG, AR-PNGase F IVIG, and AR-CD28 groups (all P<0.01), with IVIG showing the better efficacy than PNGase F IVIG. Furthermore, blood and spleen FcγRIA and FcγRIIIA were increased by AR, while FcγRIIB expressions in splenic activated B cells and regulatory B cells were decreased; these changes were partly alleviated by all three treatments, with IVIG having the better effect than PNGase F IVIG. CONCLUSION We observed an association between B cell FcγRIIB expression and treatment efficacy for AR after kidney transplantation in rats.
Collapse
Affiliation(s)
- Juan Jin
- Department of Nephrology, Zhejiang Provincial People's Hospital, Zhejiang 310014, PR China; People's Hospital of Hangzhou Medical College, Zhejiang 310014, PR China
| | - Jianguang Gong
- Department of Nephrology, Zhejiang Provincial People's Hospital, Zhejiang 310014, PR China; People's Hospital of Hangzhou Medical College, Zhejiang 310014, PR China
| | - Bo Lin
- Department of Nephrology, Zhejiang Provincial People's Hospital, Zhejiang 310014, PR China; People's Hospital of Hangzhou Medical College, Zhejiang 310014, PR China
| | - Yiwen Li
- Department of Nephrology, Zhejiang Provincial People's Hospital, Zhejiang 310014, PR China; People's Hospital of Hangzhou Medical College, Zhejiang 310014, PR China
| | - Qiang He
- Department of Nephrology, Zhejiang Provincial People's Hospital, Zhejiang 310014, PR China; People's Hospital of Hangzhou Medical College, Zhejiang 310014, PR China.
| |
Collapse
|
43
|
Benichou G, Gonzalez B, Marino J, Ayasoufi K, Valujskikh A. Role of Memory T Cells in Allograft Rejection and Tolerance. Front Immunol 2017; 8:170. [PMID: 28293238 PMCID: PMC5328996 DOI: 10.3389/fimmu.2017.00170] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 02/02/2017] [Indexed: 12/30/2022] Open
Abstract
Memory T cells are characterized by their low activation threshold, robust effector functions, and resistance to conventional immunosuppression and costimulation blockade. Unlike their naïve counterparts, memory T cells reside in and recirculate through peripheral non-lymphoid tissues. Alloreactive memory T cells are subdivided into different categories based on their origins, phenotypes, and functions. Recipients whose immune systems have been directly exposed to allogeneic major histocompatibility complex (MHC) molecules display high affinity alloreactive memory T cells. In the absence of any prior exposure to allogeneic MHC molecules, endogenous alloreactive memory T cells are regularly generated through microbial infections (heterologous immunity). Regardless of their origin, alloreactive memory T cells represent an essential element of the allograft rejection process and a major barrier to tolerance induction in clinical transplantation. This article describes the different subsets of alloreactive memory T cells involved in transplant rejection and examine their generation, functional properties, and mechanisms of action. In addition, we discuss strategies developed to target deleterious allospecific memory T cells in experimental animal models and clinical settings.
Collapse
Affiliation(s)
- Gilles Benichou
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Bruno Gonzalez
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jose Marino
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Katayoun Ayasoufi
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Anna Valujskikh
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
44
|
Poirier N, Blancho G, Hiance M, Mary C, Van Assche T, Lempoels J, Ramael S, Wang W, Thepenier V, Braudeau C, Salabert N, Josien R, Anderson I, Gourley I, Soulillou JP, Coquoz D, Vanhove B. First-in-Human Study in Healthy Subjects with FR104, a Pegylated Monoclonal Antibody Fragment Antagonist of CD28. THE JOURNAL OF IMMUNOLOGY 2016; 197:4593-4602. [PMID: 27849166 DOI: 10.4049/jimmunol.1601538] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 10/13/2016] [Indexed: 01/03/2023]
Abstract
FR104 is a monovalent pegylated Fab' Ab, antagonist of CD28, under development for treatment of transplant rejection and autoimmune diseases. In contrast to CD80/86 antagonists (CTLA4-Ig), FR104 selectively blunts CD28 costimulation while sparing CTLA-4 and PD-L1 coinhibitory signals. In the present work, FR104 has been evaluated in a first-in-human study to evaluate the safety, pharmacokinetics, pharmacodynamics, and potency of i.v. administrations in healthy subjects. Sixty-four subjects were randomly assigned to four single ascending dose groups, two double dose groups and four single ascending dose groups challenged with keyhole limpet hemocyanin. Subjects were followed up over a maximum of 113 d. Overall, the pharmacokinetics of FR104 after a single and double infusions was approximately linear at doses ≥0.200 mg/kg. CD28 receptor occupancy by FR104 was saturated at the first sampling time point (0.5 h) at doses above 0.02 mg/kg and returned to 50% in a dose-dependent manner, by day 15 (0.020 mg/kg) to 85 (1.500 mg/kg). FR104 was well tolerated, with no evidence of cytokine-release syndrome and no impact on blood lymphocyte subsets. Inhibition of anti-keyhole limpet hemocyanin Ab response was dose-dependent in FR104 recipients and was already apparent at a dose of 0.02 mg/kg. Abs to FR104 were detected in 22/46 (48%) of FR104 recipients and only 1/46 (2.2%) was detected during drug exposure. In conclusion, selective blockade of CD28 with FR104 was safe and well tolerated at the doses tested. The observed immunosuppressive activity indicated that FR104 has potential to show clinical activity in the treatment of immune-mediated diseases.
Collapse
Affiliation(s)
- Nicolas Poirier
- Institut National de la Santé et de la Recherche Médicale UMR 1064, Nantes F44093, France.,Institut de Transplantation Urologie Néphrologie, Centre Hospitalier Universitaire, Université de Nantes, Nantes F44000, France.,OSE Immunotherapeutics S.A., Nantes F44200, France
| | - Gilles Blancho
- Institut National de la Santé et de la Recherche Médicale UMR 1064, Nantes F44093, France.,Institut de Transplantation Urologie Néphrologie, Centre Hospitalier Universitaire, Université de Nantes, Nantes F44000, France.,Centre Hospitalier Universitaire Nantes, Laboratoire d'Immunologie, Centre d'immunomonitorage Nantes-Atlantique, Nantes, F44000, France
| | | | - Caroline Mary
- Institut National de la Santé et de la Recherche Médicale UMR 1064, Nantes F44093, France.,Institut de Transplantation Urologie Néphrologie, Centre Hospitalier Universitaire, Université de Nantes, Nantes F44000, France.,OSE Immunotherapeutics S.A., Nantes F44200, France
| | - Tim Van Assche
- SGS Life Science Services, Clinical Pharmacology Unit Antwerp, Antwerp 2060, Belgium
| | - Jos Lempoels
- SGS Life Science Services, Clinical Pharmacology Unit Antwerp, Antwerp 2060, Belgium
| | - Steven Ramael
- SGS Life Science Services, Clinical Pharmacology Unit Antwerp, Antwerp 2060, Belgium
| | - Weirong Wang
- Janssen Research & Development, LLC, Spring House, PA 19477
| | - Virginie Thepenier
- Institut National de la Santé et de la Recherche Médicale UMR 1064, Nantes F44093, France.,Institut de Transplantation Urologie Néphrologie, Centre Hospitalier Universitaire, Université de Nantes, Nantes F44000, France.,OSE Immunotherapeutics S.A., Nantes F44200, France
| | - Cecile Braudeau
- Institut National de la Santé et de la Recherche Médicale UMR 1064, Nantes F44093, France.,Institut de Transplantation Urologie Néphrologie, Centre Hospitalier Universitaire, Université de Nantes, Nantes F44000, France.,LabEx ImmunoGraft Oncology, Nantes F44000, Nantes, France; and
| | - Nina Salabert
- Institut National de la Santé et de la Recherche Médicale UMR 1064, Nantes F44093, France.,Institut de Transplantation Urologie Néphrologie, Centre Hospitalier Universitaire, Université de Nantes, Nantes F44000, France.,Centre Hospitalier Universitaire Nantes, Laboratoire d'Immunologie, Centre d'immunomonitorage Nantes-Atlantique, Nantes, F44000, France.,LabEx ImmunoGraft Oncology, Nantes F44000, Nantes, France; and
| | - Regis Josien
- Institut National de la Santé et de la Recherche Médicale UMR 1064, Nantes F44093, France.,Institut de Transplantation Urologie Néphrologie, Centre Hospitalier Universitaire, Université de Nantes, Nantes F44000, France.,Centre Hospitalier Universitaire Nantes, Laboratoire d'Immunologie, Centre d'immunomonitorage Nantes-Atlantique, Nantes, F44000, France.,LabEx ImmunoGraft Oncology, Nantes F44000, Nantes, France; and
| | - Ian Anderson
- Janssen Research & Development, LLC, Spring House, PA 19477
| | - Ian Gourley
- Janssen Research & Development, LLC, Spring House, PA 19477
| | - Jean-Paul Soulillou
- Institut National de la Santé et de la Recherche Médicale UMR 1064, Nantes F44093, France.,Institut de Transplantation Urologie Néphrologie, Centre Hospitalier Universitaire, Université de Nantes, Nantes F44000, France
| | | | - Bernard Vanhove
- Institut National de la Santé et de la Recherche Médicale UMR 1064, Nantes F44093, France; .,Institut de Transplantation Urologie Néphrologie, Centre Hospitalier Universitaire, Université de Nantes, Nantes F44000, France.,OSE Immunotherapeutics S.A., Nantes F44200, France.,LabEx ImmunoGraft Oncology, Nantes F44000, Nantes, France; and
| |
Collapse
|
45
|
Malvezzi P, Jouve T, Rostaing L. Costimulation Blockade in Kidney Transplantation: An Update. Transplantation 2016; 100:2315-2323. [PMID: 27472094 PMCID: PMC5084636 DOI: 10.1097/tp.0000000000001344] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/04/2016] [Accepted: 05/10/2016] [Indexed: 12/15/2022]
Abstract
In the setting of solid-organ transplantation, calcineurin inhibitor (CNI)-based therapy remains the cornerstone of immunosuppression. However, long-term use of CNIs is associated with some degree of nephrotoxicity. This has led to exploring the blockade of some costimulation pathways as an efficient immunosuppressive tool instead of using CNIs. The only agent already in clinical use and approved by the health authorities for kidney transplant patients is belatacept (Nulojix), a fusion protein that interferes with cytotoxic T lymphocyte-associated protein 4. Belatacept has been demonstrated to be as efficient as cyclosporine-based immunosuppression and is associated with significantly better renal function, that is, no nephrotoxicity. However, in the immediate posttransplant period, significantly more mild/moderate episodes of acute rejection have been reported, favored by the fact that cytotoxic T lymphocyte-associated protein pathway has an inhibitory effect on the alloimmune response; thereby its inhibition is detrimental in this regard. This has led to the development of antibodies that target CD28. The most advanced is FR104, it has shown promise in nonhuman primate models of autoimmune diseases and allotransplantation. In addition, research into blocking the CD40-CD154 pathway is underway. A phase II study testing ASK1240, that is, anti-CD40 antibody has been completed, and the results are pending.
Collapse
Affiliation(s)
- Paolo Malvezzi
- Clinique Universitaire de Néphrologie, Unité de Transplantation Rénale, CHU Grenoble, France
| | - Thomas Jouve
- Clinique Universitaire de Néphrologie, Unité de Transplantation Rénale, CHU Grenoble, France
- Université Joseph Fourier, Grenoble, France
| | - Lionel Rostaing
- Clinique Universitaire de Néphrologie, Unité de Transplantation Rénale, CHU Grenoble, France
- UniversitéToulouse III Paul Sabatier, Toulouse, France
- INSERM U563, IFR-BMT, CHU Purpan, Toulouse, France
| |
Collapse
|
46
|
Abstract
Immunosuppression strategies that selectively inhibit effector T cells while preserving and even enhancing CD4FOXP3 regulatory T cells (Treg) permit immune self-regulation and may allow minimization of immunosuppression and associated toxicities. Many immunosuppressive drugs were developed before the identity and function of Treg were appreciated. A good understanding of the interactions between Treg and immunosuppressive agents will be valuable to the effective design of more tolerable immunosuppression regimens. This review will discuss preclinical and clinical evidence regarding the influence of current and emerging immunosuppressive drugs on Treg homeostasis, stability, and function as a guideline for the selection and development of Treg-friendly immunosuppressive regimens.
Collapse
Affiliation(s)
- Akiko Furukawa
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Steven A Wisel
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
47
|
Monaco S, Jahraus B, Samstag Y, Bading H. Nuclear calcium is required for human T cell activation. J Cell Biol 2016; 215:231-243. [PMID: 27810914 PMCID: PMC5084645 DOI: 10.1083/jcb.201602001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 09/27/2016] [Indexed: 01/12/2023] Open
Abstract
Monaco et al. demonstrate that calcium signals in activated human T cells consist of a cytoplasmic and a nuclear component, which are both required for the immune response. Blockade of nuclear calcium signaling inhibits T cell activation and induces an anergy-like state. Calcium signals in stimulated T cells are generally considered single entities that merely trigger immune responses, whereas costimulatory events specify the type of reaction. Here we show that the “T cell calcium signal” is a composite signal harboring two distinct components that antagonistically control genomic programs underlying the immune response. Using human T cells from healthy individuals, we establish nuclear calcium as a key signal in human T cell adaptogenomics that drives T cell activation and is required for signaling to cyclic adenosine monophosphate response element–binding protein and the induction of CD25, CD69, interleukin-2, and γ-interferon. In the absence of nuclear calcium signaling, cytosolic calcium activating nuclear factor of activated T cells translocation directed the genomic response toward enhanced expression of genes that negatively modulate T cell activation and are associated with a hyporesponsive state. Thus, nuclear calcium controls the T cell fate decision between a proliferative immune response and tolerance. Modulators of nuclear calcium–driven transcription may be used to develop a new type of pro-tolerance immunosuppressive therapy.
Collapse
Affiliation(s)
- Sara Monaco
- Interdisciplinary Center for Neurosciences, Department of Neurobiology, Heidelberg University, 69120 Heidelberg, Germany
| | - Beate Jahraus
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, 69120 Heidelberg, Germany
| | - Yvonne Samstag
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, 69120 Heidelberg, Germany
| | - Hilmar Bading
- Interdisciplinary Center for Neurosciences, Department of Neurobiology, Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
48
|
Herz S, Höfer T, Papapanagiotou M, Leyh JC, Meyenburg S, Schadendorf D, Ugurel S, Roesch A, Livingstone E, Schilling B, Franklin C. Checkpoint inhibitors in chronic kidney failure and an organ transplant recipient. Eur J Cancer 2016; 67:66-72. [PMID: 27614165 DOI: 10.1016/j.ejca.2016.07.026] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 07/28/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND Immune-checkpoint inhibitors have been approved for the treatment of metastatic melanoma based on several phase III trials. Patients after organ transplantation and patients with impaired renal function were excluded from these studies. Recently, allograft rejections were reported in organ transplant recipients receiving PD-1 blocking antibodies. PATIENTS AND FINDINGS Four patients with metastatic melanoma and impaired kidney function (baseline serum creatinine 1.79-2.59 mg/dl) were treated with immune-checkpoint blockers, of which one was a kidney-transplant recipient receiving immunosuppressive therapy with tacrolimus and prednisolone. The patient was initially treated with the anti-CTLA-4 antibody ipilimumab after detailed explanation of the potential risk of allograft rejection. Upon disease progression, therapy was switched to the anti-PD-1 antibody nivolumab. The other three patients were treated with nivolumab or pembrolizumab, two of them after previous therapy with ipilimumab. RESULTS The patients received a median of six doses (range 3-21) of anti-PD-1 antibodies and 3-4 doses of ipilimumab. Kidney function tests remained stable throughout the course of checkpoint blockade. In the kidney transplant recipient, neither ipilimumab nor nivolumab led to an allograft rejection. Responses to anti-PD-1 treatment were divergent with two patients showing disease progression, one achieving a mixed response and one experiencing a complete response. CONCLUSION These cases show that checkpoint inhibitors can be a safe therapeutic option in patients with impaired kidney function. Furthermore, we report the first organ transplant patient with malignant melanoma who received ipilimumab followed by nivolumab without experiencing a kidney allograft rejection.
Collapse
Affiliation(s)
- Saskia Herz
- Department of Dermatology, Venereology and Allergology, University Hospital, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Thomas Höfer
- Department of Dermatology, Venereology and Allergology, University Hospital, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Matina Papapanagiotou
- Department of Dermatology, Venereology and Allergology, University Hospital, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Julia Christina Leyh
- Department of Dermatology, Venereology and Allergology, University Hospital, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Sarah Meyenburg
- Department of Dermatology, Venereology and Allergology, University Hospital, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Dirk Schadendorf
- Department of Dermatology, Venereology and Allergology, University Hospital, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Selma Ugurel
- Department of Dermatology, Venereology and Allergology, University Hospital, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Alexander Roesch
- Department of Dermatology, Venereology and Allergology, University Hospital, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Elisabeth Livingstone
- Department of Dermatology, Venereology and Allergology, University Hospital, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Bastian Schilling
- Department of Dermatology, Venereology and Allergology, University Hospital, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany.
| | - Cindy Franklin
- Department of Dermatology, Venereology and Allergology, University Hospital, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW Transplantation tolerance, successful acceptance of an organ without the perils of immunosuppression, has been a central goal of transplant research. Many strategies to achieve this tolerance have been examined over the past three decades, culminating in several human trials of transplant tolerance. This progression from the 'benchtop to the clinic' has depended on the successful implementation of these tolerance strategies in nonhuman primates. This review will examine the described methods of transplant tolerance induction in nonhuman primates. RECENT FINDINGS Although costimulatory blockade and mixed chimerism have an established record of achieving transplant tolerance in nonhuman primates, some of the most innovative recent techniques of tolerance induction have relied on cellular transfer. This review will fully examine the role of regulatory T-cell transfer and the use of mesenchymal stem/stromal cells to promote tolerance of organ allografts in nonhuman primates. SUMMARY Use of translational nonhuman primate transplant models is a vital intermediate step to advance new approaches of transplant tolerance induction from the lab to the clinic. This review will explore numerous techniques of tolerance induction that have been piloted in primates, including depletional techniques, induction of mixed hematopoietic chimerism, costimulation blockade, and adoptive transfer of tolerogenic cell populations.
Collapse
|
50
|
Abstract
The ultimate outcome of alloreactivity versus tolerance following transplantation is potently influenced by the constellation of cosignaling molecules expressed by immune cells during priming with alloantigen, and the net sum of costimulatory and coinhibitory signals transmitted via ligation of these molecules. Intense investigation over the last two decades has yielded a detailed understanding of the kinetics, cellular distribution, and intracellular signaling networks of cosignaling molecules such as the CD28, TNF, and TIM families of receptors in alloimmunity. More recent work has better defined the cellular and molecular mechanisms by which engagement of cosignaling networks serve to either dampen or augment alloimmunity. These findings will likely aid in the rational development of novel immunomodulatory strategies to prolong graft survival and improve outcomes following transplantation.
Collapse
Affiliation(s)
- Mandy L Ford
- Emory Transplant Center and Department of Surgery, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|