1
|
Mohanty S, Barik P, Debata N, Nagarajan P, Devadas S. iCa 2+ Flux, ROS and IL-10 Determines Cytotoxic, and Suppressor T Cell Functions in Chronic Human Viral Infections. Front Immunol 2020; 11:83. [PMID: 32210950 PMCID: PMC7068714 DOI: 10.3389/fimmu.2020.00083] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 01/13/2020] [Indexed: 12/13/2022] Open
Abstract
Exhaustion of CD8+ T cells and increased IL-10 production is well-known in chronic viral infections but mechanisms leading to loss of their cytotoxic capabilities and consequent exhaustion remain unclear. Exhausted CD8+T cells also called T suppressors are highly immune suppressive with altered T cell receptor signaling characteristics that mark it exclusively from their cytotoxic counterparts. Our study found that iCa2+ flux is reduced following T cell receptor activation in T suppressor cells when compared to their effector counterpart. Importantly chronic activation of murine cytotoxic CD8+ T cells lead to reduced iCa2+ influx, decreased IFN-γ and enhanced IL-10 production and this profile is mimicked in Tc1 cells upon reduction of iCa2+ flux by extracellular calcium channel inhibitors. Further reduced iCa2+ flux induced ROS which lead to IFN-γ reduction and increased IL-10 producing T suppressors through the STAT3—STAT5 axis. The above findings were substantiated by our human data where reduced iCa2+ flux in chronic Hepatitis infections displayed CD8+ T cells with low IFN-γ and increased IL-10 production. Importantly treatment with an antioxidant led to increased IFN-γ and reduced IL-10 production in human chronic Hep-B/C samples suggesting overall a proximal regulatory role for iCa2+ influx, ROS, and IL-10 in determining the effector/ suppressive axis of CD8+ T cells.
Collapse
Affiliation(s)
- Subhasmita Mohanty
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, India
| | - Prakash Barik
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, India
| | - Nagen Debata
- Department of Pathology, Institute of Medical Sciences and SUM Hospital, Bhubaneswar, India
| | - Perumal Nagarajan
- Experimental Animal Facility, National Institute of Immunology, New Delhi, India
| | - Satish Devadas
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, India
| |
Collapse
|
2
|
Kumar S, Leigh ND, Cao X. The Role of Co-stimulatory/Co-inhibitory Signals in Graft-vs.-Host Disease. Front Immunol 2018; 9:3003. [PMID: 30627129 PMCID: PMC6309815 DOI: 10.3389/fimmu.2018.03003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 12/05/2018] [Indexed: 12/31/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is an effective immunotherapeutic approach for various hematologic and immunologic ailments. Despite the beneficial impact of allo-HCT, its adverse effects cause severe health concerns. After transplantation, recognition of host cells as foreign entities by donor T cells induces graft-vs.-host disease (GVHD). Activation, proliferation and trafficking of donor T cells to target organs and tissues are critical steps in the pathogenesis of GVHD. T cell activation is a synergistic process of T cell receptor (TCR) recognition of major histocompatibility complex (MHC)-anchored antigen and co-stimulatory/co-inhibitory signaling in the presence of cytokines. Most of the currently used therapeutic regimens for GVHD are based on inhibiting the allogeneic T cell response or T-cell depletion (TCD). However, the immunosuppressive drugs and TCD hamper the therapeutic potential of allo-HCT, resulting in attenuated graft-vs.-leukemia (GVL) effect as well as increased vulnerability to infection. In view of the drawback of overbroad immunosuppression, co-stimulatory, and co-inhibitory molecules are plausible targets for selective modulation of T cell activation and function that can improve the effectiveness of allo-HCT. Therefore, this review collates existing knowledge of T cell co-stimulation and co-inhibition with current research that may have the potential to provide novel approaches to cure GVHD without sacrificing the beneficial effects of allo-HCT.
Collapse
Affiliation(s)
- Sandeep Kumar
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Nicholas D Leigh
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Xuefang Cao
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States.,Department of Microbiology and Immunology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
3
|
Du W, Cao X. Cytotoxic Pathways in Allogeneic Hematopoietic Cell Transplantation. Front Immunol 2018; 9:2979. [PMID: 30631325 PMCID: PMC6315278 DOI: 10.3389/fimmu.2018.02979] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/04/2018] [Indexed: 12/11/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is a potentially curative treatment for hematologic malignancies, and other hematologic and immunologic diseases. Donor-derived immune cells identify and attack cancer cells in the patient producing a unique graft-vs.-tumor (GVT) effect. This beneficial response renders allo-HCT one of the most effective forms of tumor immunotherapy. However, alloreactive donor T cells can damage normal host cells thereby causing graft-vs.-host disease (GVHD), which results in substantial morbidity and mortality. To date, GVHD remains as the major obstacle for more successful application of allo-HCT. Of special significance in this context are a number of cytotoxic pathways that are involved in GVHD and GVT response as well as donor cell engraftment. In this review, we summarize progress in the investigation of these cytotoxic pathways, including Fas/Fas ligand (FasL), perforin/granzyme, and cytokine pathways. Many studies have delineated their distinct operating mechanisms and how they are involved in the complex cellular interactions amongst donor, host, tumor, and infectious pathogens. Driven by progressing elucidation of their contributions in immune reconstitution and regulation, various interventional strategies targeting these pathways have entered translational stages with aims to improve the effectiveness of allo-HCT.
Collapse
Affiliation(s)
- Wei Du
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Xuefang Cao
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States.,Department of Microbiology and Immunology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
4
|
Ito H, Ando T, Seishima M. Inhibition of iNOS activity enhances the anti-tumor effects of alpha-galactosylceramide in established murine cancer model. Oncotarget 2016; 6:41863-74. [PMID: 26496031 PMCID: PMC4747194 DOI: 10.18632/oncotarget.6172] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 10/06/2015] [Indexed: 12/15/2022] Open
Abstract
Alpha-garactosylceramide (GalCer) has been shown to have anti-tumor effect in the basic research and clinical studies. However, anti-tumor effect of GalCer is limited. The administration of GalCer increases the production of IFN-γ which is involved in the suppression of tumor growth. On the other hand, the enhancement of IFN-γ production increases immunosuppressive factors such as nitric oxide. This suppressive action might impair the anti-tumor effect of GalCer. In the present study, we evaluated the anti-tumor effect of GalCer in the absence of inducible nitric oxide synthase (iNOS). In lung metastatic model, the number of tumor nodules in the lung of iNOS-KO mice treated with GalCer was significantly reduced compared with that of WT mice treated with GalCer. Moreover, L-NAME, which is the inhibitor for iNOS, enhanced the anti-tumor effect of GalCer in lung metastatic model. The frequency of CD8+ cells in bronchoalveolar lavage fluid increased in iNOS-KO mice treated with GalCer. The administration of GalCer increased the frequency of myeloid-derived suppressor cells (MDSCs) in the lung from tumor-bearing WT mice, but the increase of MDSCs in the lung was not induced in iNOS-KO mice. The subcutaneous tumor experiments revealed that the administration of GalCer in the absence of iNOS expression significantly enhanced the induction of tumor antigen-specific response. Finally, our results indicated that the inhibition of iNOS expression could enhance the therapeutic efficacy of GalCer via the increase of tumor antigen-specific immune response and the suppression of MDSCs.
Collapse
Affiliation(s)
- Hiroyasu Ito
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Yanagido, Gifu, Japan
| | - Tatsuya Ando
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Yanagido, Gifu, Japan
| | - Mitsuru Seishima
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Yanagido, Gifu, Japan
| |
Collapse
|
5
|
Leignadier J, Favre S, Luther SA, Luescher IF. CD8 engineered cytotoxic T cells reprogram melanoma tumor environment. Oncoimmunology 2015; 5:e1086861. [PMID: 27141342 DOI: 10.1080/2162402x.2015.1086861] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 08/18/2015] [Accepted: 08/20/2015] [Indexed: 10/23/2022] Open
Abstract
Cytotoxic T lymphocytes (CTL) from CD8β-deficient mice have powerful FasL-mediated cytotoxicity and IFNγ responses, but ablated Ca2+ and NFAT signaling, which can be restored by transduction with CD8β. Upon infection with lymphocytic choriomeningitis virus (LCMV), these cells yielded GP33-specific CTL (CD8βR) that exhibited high FasL/Fas-mediated cytotoxicity, IFNγ CXCL9 and 10 chemokine responses. Transfer of these cells in B16-GP33 tumor bearing mice resulted in (i) massive T cell tumor infiltration, (ii) strong reduction of myeloid-derived suppressor cells (MDSCs), regulatory T cells (Treg) and IL-17-expressing T helper cells, (iii) maturation of tumor-associated antigen-presenting cells and (iv) production of endogenous, B16 melanoma-specific CTL that eradicated the tumor long after the transferred CD8βR CTL perished. Our study demonstrates that the synergistic combination of strong Fas/FasL mediated cytotoxicity, IFNγ and CXCL9 and 10 responses endows adoptively transferred CTL to reprogram the tumor environment and to thus enable the generation of endogenous, tumoricidal immunity.
Collapse
Affiliation(s)
- Julie Leignadier
- Ludwig Center for Cancer Research, University of Lausanne , Epalinges, Switzerland
| | - Stephanie Favre
- Department of Biochemistry, University of Lausanne , Epalinges, Switzerland
| | - Sanjiv A Luther
- Department of Biochemistry, University of Lausanne , Epalinges, Switzerland
| | - Immanuel F Luescher
- Ludwig Center for Cancer Research, University of Lausanne , Epalinges, Switzerland
| |
Collapse
|
6
|
Mossoba ME, Halverson DC, Kurlander R, Schuver BB, Carpenter A, Hansen B, Steinberg SM, Ali SA, Tageja N, Hakim FT, Gea-Banacloche J, Sportes C, Hardy NM, Hickstein DD, Pavletic SZ, Khuu H, Sabatini M, Stroncek D, Levine BL, June CH, Mariotti J, Rixe O, Fojo AT, Bishop MR, Gress RE, Fowler DH. High-Dose Sirolimus and Immune-Selective Pentostatin plus Cyclophosphamide Conditioning Yields Stable Mixed Chimerism and Insufficient Graft-versus-Tumor Responses. Clin Cancer Res 2015; 21:4312-20. [PMID: 26071480 DOI: 10.1158/1078-0432.ccr-15-0340] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 05/26/2015] [Indexed: 02/07/2023]
Abstract
PURPOSE We hypothesized that lymphoid-selective host conditioning and subsequent adoptive transfer of sirolimus-resistant allogeneic T cells (T-Rapa), when combined with high-dose sirolimus drug therapy in vivo, would safely achieve antitumor effects while avoiding GVHD. EXPERIMENTAL DESIGN Patients (n = 10) with metastatic renal cell carcinoma (RCC) were accrued because this disease is relatively refractory to high-dose conditioning yet may respond to high-dose sirolimus. A 21-day outpatient regimen of weekly pentostatin (P; 4 mg/m(2)/dose) combined with daily, dose-adjusted cyclophosphamide (C; ≤200 mg/d) was designed to deplete and suppress host T cells. After PC conditioning, patients received matched sibling, T-cell-replete peripheral blood stem cell allografts, and high-dose sirolimus (serum trough target, 20-30 ng/mL). To augment graft-versus-tumor (GVT) effects, multiple T-Rapa donor lymphocyte infusions (DLI) were administered (days 0, 14, and 45 posttransplant), and sirolimus was discontinued early (day 60 posttransplant). RESULTS PC conditioning depleted host T cells without neutropenia or infection and facilitated donor engraftment (10 of 10 cases). High-dose sirolimus therapy inhibited multiple T-Rapa DLI, as evidenced by stable mixed donor/host chimerism. No antitumor responses were detected by RECIST criteria and no significant classical acute GVHD was observed. CONCLUSIONS Immune-selective PC conditioning represents a new approach to safely achieve alloengraftment without neutropenia. However, allogeneic T cells generated ex vivo in sirolimus are not resistant to the tolerance-inducing effects of in vivo sirolimus drug therapy, thereby cautioning against use of this intervention in patients with refractory cancer.
Collapse
Affiliation(s)
- Miriam E Mossoba
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | - David C Halverson
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Roger Kurlander
- Department of Laboratory Medicine, Clinical Center, NIH, Bethesda, Maryland
| | - Bazetta Blacklock Schuver
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Ashley Carpenter
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Brenna Hansen
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | | | - Syed Abbas Ali
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Nishant Tageja
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Frances T Hakim
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Juan Gea-Banacloche
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Claude Sportes
- Georgia Regents University Cancer Center, Augusta, Georgia
| | - Nancy M Hardy
- University of Maryland Greenbaum Cancer Center, Baltimore, Maryland
| | - Dennis D Hickstein
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Steven Z Pavletic
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Hanh Khuu
- Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, Maryland
| | - Marianna Sabatini
- Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, Maryland
| | - David Stroncek
- Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, Maryland
| | - Bruce L Levine
- University of Pennsylvania, Abramson Family Cancer Research Center, Philadelphia, Pennsylvania
| | - Carl H June
- University of Pennsylvania, Abramson Family Cancer Research Center, Philadelphia, Pennsylvania
| | - Jacopo Mariotti
- Fondazione IRCCS Instituto Nazionale dei Tumori, Milan, Italy
| | - Olivier Rixe
- University of New Mexico Cancer Center, Albuquerque, New Mexico
| | - Antonio Tito Fojo
- Genitourinary Malignancies Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | | | - Ronald E Gress
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Daniel H Fowler
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland.
| |
Collapse
|
7
|
Wang H, Yang YG. The complex and central role of interferon-γ in graft-versus-host disease and graft-versus-tumor activity. Immunol Rev 2015; 258:30-44. [PMID: 24517424 DOI: 10.1111/imr.12151] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 11/01/2013] [Accepted: 11/11/2013] [Indexed: 12/22/2022]
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is increasingly being performed to treat patients with hematologic malignancies. However, separating the beneficial graft-versus-tumor (GVT) or graft-versus-leukemia effects from graft-versus-host disease (GVHD) has been difficult and remains a significant challenge toward improving therapeutic efficacy and reducing toxicity of allo-HCT. GVHD is induced by donor T cells that also mediate potent anti-tumor responses. However, despite the largely shared effector mechanisms, extensive animal studies have demonstrated the potential of dissociating the GVT effect from GVHD. Also in many clinical cases, long-term remission was achieved following allo-HCT, without significant GVHD. A better mechanistic understanding of the immunopathophysiology of GVHD and GVT effects may potentially help to improve allo-HCT as well as maximize the benefit of GVT effects while minimizing GVHD. In this article, we review the role of IFN-γ in regulation of alloresponses following allo-HCT, with a focus on the mechanisms of how this cytokine may separate GVHD from GVT effects.
Collapse
Affiliation(s)
- Hui Wang
- Columbia Center for Translational Immunology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | | |
Collapse
|
8
|
Oelkrug C, Sack U, Boldt A, Nascimento IC, Ulrich H, Fricke S. Antibody- and aptamer-strategies for GvHD prevention. J Cell Mol Med 2014; 19:11-20. [PMID: 25353670 PMCID: PMC4288345 DOI: 10.1111/jcmm.12416] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 08/01/2014] [Indexed: 02/06/2023] Open
Abstract
Prevention of Graft-versus-Host-Disease (GvHD) by preserved Graft-versus-Leukaemia (GvL) effect is one of the major obstacles following allogeneic haematopoietic stem cell transplantation. Currently used drugs are associated with side effects and were not able to separate GvHD from the GvL-effect because of general T-cell suppression. This review focuses on murine models for GvHD and currently available treatment options involving antibodies and applications for the therapeutic use of aptamers as well as strategies for targeting immune responses by allogenic antigens.
Collapse
Affiliation(s)
- Christopher Oelkrug
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
| | | | | | | | | | | |
Collapse
|
9
|
Zhao Q, Tong L, He N, Feng G, Leng L, Sun W, Xu Y, Wang Y, Xiang R, Li Z. IFN-γ mediates graft-versus-breast cancer effects via enhancing cytotoxic T lymphocyte activity. Exp Ther Med 2014; 8:347-354. [PMID: 25009582 PMCID: PMC4079438 DOI: 10.3892/etm.2014.1760] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 05/13/2014] [Indexed: 12/11/2022] Open
Abstract
Previous studies have demonstrated the beneficial effect of graft-versus-tumor (GVT) following hematopoietic stem cell transplantation (HSCT) on the incidence of leukemia relapse and the overall survival rate of patients with leukemia; however, detailed mechanisms underlying the effects GVT exhibits on solid tumors following allogeneic HSCT are yet to be elucidated. The aim of the present study was to investigate the immune mechanism underlying the effect of interferon (IFN)-γ on GVT following allogeneic HSCT in breast cancer therapy. An in situ breast cancer mouse model was established by injecting 5×104 4T1 cells into the mammary fat pads of BALB/c mice. The 4T1 cells were transfected with the firefly luciferase reporter gene in order to monitor the tumor progression in real time. An allogeneic HSCT model was then established by transplanting bone marrow mononuclear cells from C57BL/6 mice to the BALB/c mice. To investigate the influence of T lymphocyte proliferation following allogeneic bone marrow transplantation, the levels of CD3+CD8+ cytotoxic T lymphocytes (CTLs) and CD4+CD25+ regulatory T cells were determined. In addition, IFN-γ and granzyme B expression levels in splenic lymphocytes were analyzed using flow cytometry. Allogeneic HSCT was found to significantly promote the proliferation and cytotoxicity of CTLs and suppress the growth of breast cancer. Furthermore, the secretory levels of IFN-γ and granzyme B by T cells were elevated following allogeneic HSCT. These results indicated that alloreactive T cells increased the secretion of IFN-γ, which promoted the alloresponse of donor CTLs. In addition, the CTLs produced granzyme B, which exerted a tumor suppressive effect.
Collapse
Affiliation(s)
- Qianjie Zhao
- Department of Pathophysiology, School of Medicine, Nankai University, Ministry of Education, Tianjin 300071, P.R. China ; Key Laboratory of Bioactive Materials, College of Life Science, Nankai University, Ministry of Education, Tianjin 300071, P.R. China
| | - Lingling Tong
- Department of Pathophysiology, School of Medicine, Nankai University, Ministry of Education, Tianjin 300071, P.R. China
| | - Ningning He
- Department of Pathophysiology, School of Medicine, Nankai University, Ministry of Education, Tianjin 300071, P.R. China
| | - Guowei Feng
- Department of Pathophysiology, School of Medicine, Nankai University, Ministry of Education, Tianjin 300071, P.R. China
| | - Liang Leng
- Department of Pathophysiology, School of Medicine, Nankai University, Ministry of Education, Tianjin 300071, P.R. China
| | - Weijun Sun
- Department of Pathophysiology, School of Medicine, Nankai University, Ministry of Education, Tianjin 300071, P.R. China
| | - Yang Xu
- Department of Pathophysiology, School of Medicine, Nankai University, Ministry of Education, Tianjin 300071, P.R. China
| | - Yuebing Wang
- Department of Pathophysiology, School of Medicine, Nankai University, Ministry of Education, Tianjin 300071, P.R. China
| | - Rong Xiang
- Department of Pathophysiology, School of Medicine, Nankai University, Ministry of Education, Tianjin 300071, P.R. China
| | - Zongjin Li
- Department of Pathophysiology, School of Medicine, Nankai University, Ministry of Education, Tianjin 300071, P.R. China ; Key Laboratory of Bioactive Materials, College of Life Science, Nankai University, Ministry of Education, Tianjin 300071, P.R. China
| |
Collapse
|
10
|
Ghosh A, Dogan Y, Moroz M, Holland AM, Yim NL, Rao UK, Young LF, Tannenbaum D, Masih D, Velardi E, Tsai JJ, Jenq RR, Penack O, Hanash AM, Smith OM, Piersanti K, Lezcano C, Murphy GF, Liu C, Palomba ML, Sauer MG, Sadelain M, Ponomarev V, van den Brink MRM. Adoptively transferred TRAIL+ T cells suppress GVHD and augment antitumor activity. J Clin Invest 2013; 123:2654-62. [PMID: 23676461 DOI: 10.1172/jci66301] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 03/14/2013] [Indexed: 01/24/2023] Open
Abstract
Current strategies to suppress graft-versus-host disease (GVHD) also compromise graft-versus-tumor (GVT) responses. Furthermore, most experimental strategies to separate GVHD and GVT responses merely spare GVT function without actually enhancing it. We have previously shown that endogenously expressed TNF-related apoptosis-inducing ligand (TRAIL) is required for optimal GVT activity against certain malignancies in recipients of allogeneic hematopoietic stem cell transplantation (allo-HSCT). In order to model a donor-derived cellular therapy, we genetically engineered T cells to overexpress TRAIL and adoptively transferred donor-type unsorted TRAIL+ T cells into mouse models of allo-HSCT. We found that murine TRAIL+ T cells induced apoptosis of alloreactive T cells, thereby reducing GVHD in a DR5-dependent manner. Furthermore, murine TRAIL+ T cells mediated enhanced in vitro and in vivo antilymphoma GVT response. Moreover, human TRAIL+ T cells mediated enhanced in vitro cytotoxicity against both human leukemia cell lines and against freshly isolated chronic lymphocytic leukemia (CLL) cells. Finally, as a model of off-the-shelf, donor-unrestricted antitumor cellular therapy, in vitro-generated TRAIL+ precursor T cells from third-party donors also mediated enhanced GVT response in the absence of GVHD. These data indicate that TRAIL-overexpressing donor T cells could potentially enhance the curative potential of allo-HSCT by increasing GVT response and suppressing GVHD.
Collapse
Affiliation(s)
- Arnab Ghosh
- Department of Immunology and Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Haploidentical hematopoietic SCT increases graft-versus-tumor effect against renal cell carcinoma. Bone Marrow Transplant 2013; 48:1084-90. [PMID: 23435516 DOI: 10.1038/bmt.2013.9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 01/10/2013] [Accepted: 01/11/2013] [Indexed: 11/08/2022]
Abstract
Allogeneic hematopoietic SCT (HSCT) has been shown to be an effective treatment option for advanced renal cell cancer (RCC). However, tumor resistance/relapse remains as the main post transplant issue. Therefore, enhancing graft-versus-tumor (GVT) activity without increasing GVHD is critical for improving the outcome of HSCT. We explored the GVT effect of haploidentical-SCT (haplo-SCT) against RCC in murine models. Lethally irradiated CB6F1 (H2K(b/d)) recipients were transplanted with T-cell-depleted BM cells from B6CBAF1 (H2K(b/k)) mice. Haplo-SCT combined with a low-dose haploidentical (HI) T-cell infusion (1 × 10(5)) successfully provided GVT activity without incurring GVHD. This effect elicited murine RCC growth control and consequently displayed a comparative survival advantage of haplo-SCT recipients when compared with MHC-matched (B6D2F1CB6F1) and parent-F1 (B6CB6F1) transplant recipients. Recipients of haplo-SCT had an increase in donor-derived splenic T-cell numbers, T-cell proliferation and IFN-γ-secreting donor-derived T-cells, a critical aspect for anti-tumor activity. The splenocytes from B6CBAF1 mice had a higher cytotoxicity against RENCA cells than the splenocytes from B6 and B6D2F1 donors after tumor challenge. These findings suggest that haplo-SCT might be an innovative immunotherapeutic platform for solid tumors, particularly for renal cell carcinoma.
Collapse
|
12
|
Bian G, Ding X, Leigh ND, Tang Y, Capitano ML, Qiu J, McCarthy PL, Liu H, Cao X. Granzyme B–Mediated Damage of CD8+T Cells Impairs Graft-versus-Tumor Effect. THE JOURNAL OF IMMUNOLOGY 2012; 190:1341-50. [DOI: 10.4049/jimmunol.1201554] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
13
|
Recombinant CD95-Fc (APG101) prevents graft-versus-host disease in mice without disabling antitumor cytotoxicity and T-cell functions. Blood 2012. [PMID: 23203823 DOI: 10.1182/blood-2012-04-423392] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Graft-versus-host disease (GVHD) induced by transplant-derived T cells represents a major complication after allogeneic bone marrow transplantation (BMT). However, these T cells support engraftment, early T-cell immunity, and mediate the graft-versus-tumor (GVT) effect. Cytotoxic effector functions by transplanted T cells are predominantly mediated by the perforin/granzyme and the CD95/CD95L system. APG101, a novel recombinant human fusion protein consisting of the extracellular domain of CD95 and the Fc domain of an IgG1 antibody inhibited CD95L-induced apoptosis without interfering with T-cell function in vitro and was therefore tested for its ability to prevent GVHD in murine BMT models across minor or major histocompatibility barriers. Starting APG101 treatment either 1 day before or 6 days after transplantation effectively reduced clinical GVHD and rescued survival between 60% and 100% if GVHD was CD95L mediated. APG101 did not interfere with the GVT effect, because P815 mastocytoma and most importantly primary Bcr-Abl-transformed B-cell leukemias were completely eradicated by the alloantigen-specific T cells. Phenotype and homing of alloantigen-specific T cells or their perforin/granzyme-mediated cytotoxicity and proliferative capacity were not affected by APG101 treatment suggesting that APG101 therapy might be useful in GVHD prophylaxis without impairing T-cell function and most importantly preserving GVT activity.
Collapse
|
14
|
Sinkovics JG. Antileukemia and antitumor effects of the graft-versus-host disease: a new immunovirological approach. Acta Microbiol Immunol Hung 2010; 57:253-347. [PMID: 21183421 DOI: 10.1556/amicr.57.2010.4.2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In leukemic mice, the native host's explicit and well-defined immune reactions to the leukemia virus (a strong exogenous antigen) and to leukemia cells (pretending in their native hosts to be protected "self" elements) are extinguished and replaced in GvHD (graft-versus-host disease) by those of the immunocompetent donor cells. In many cases, the GvHD-inducer donors display genetically encoded resistance to the leukemia virus. In human patients only antileukemia and anti-tumor cell immune reactions are mobilized; thus, patients are deprived of immune reactions to a strong exogenous antigen (the elusive human leukemia-sarcoma retroviruses). The innate and adaptive immune systems of mice have to sustain the immunosuppressive effects of leukemia-inducing retroviruses. Human patients due to the lack of leukemiainducing retroviral pathogens (if they exist, they have not as yet been discovered), escape such immunological downgrading. After studying leukemogenic retroviruses in murine and feline (and other mammalian) hosts, it is very difficult to dismiss retroviral etiology for human leukemias and sarcomas. Since no characterized and thus recognized leukemogenic-sarcomagenic retroviral agents are being isolated from the vast majority of human leukemias-sarcomas, the treatment for these conditions in mice and in human patients vastly differ. It is immunological and biological modalities (alpha interferons; vaccines; adoptive lymphocyte therapy) that dominate the treatment of murine leukemias, whereas combination chemotherapy remains the main remission-inducing agent in human leukemias-lymphomas and sarcomas (as humanized monoclonal antibodies and immunotoxins move in). Yet, in this apparently different backgrounds in Mus and Homo, GvHD, as a treatment modality, appears to work well in both hosts, by replacing the hosts' anti-leukemia and anti-tumor immune faculties with those of the donor. The clinical application of GvHD in the treatment of human leukemias-lymphomas and malignant solid tumors remains a force worthy of pursuit, refinement and strengthening. Graft engineering and modifications of the inner immunological environment of the recipient host by the activation or administration of tumor memory T cells, selected Treg cells and natural killer (NKT) cell classes and cytokines, and the improved pharmacotherapy of GvHD without reducing its antitumor efficacy, will raise the value of GvHD to the higher ranks of the effective antitumor immunotherapeutical measures. Clinical interventions of HCT/HSCT (hematopoietic cell/stem cell transplants) are now applicable to an extended spectrum of malignant diseases in human patients, being available to elderly patients, who receive non-myeloablative conditioning, are re-enforced by post-transplant donor lymphocyte (NK cell and immune T cell) infusions and post-transplant vaccinations, and the donor cells may derive from engineered grafts, or from cord blood with reduced GvHD, but increased GvL/GvT-inducing capabilities (graft-versus leukemia/tumor). Post-transplant T cell transfusions are possible only if selected leukemia antigen-specific T cell clones are available. In verbatim quotation: "Ultimately, advances in separation of GvT from GvHD will further enhance the potential of allogeneic HCT as a curative treatment for hematological malignancies" (Rezvani, A.R. and Storb, R.F., Journal of Autoimmunity 30:172-179, 2008 (see in the text)). It may be added: for cure, a combination of the GvL/T effects with new targeted therapeutic modalities, as elaborated on in this article, will be necessary.
Collapse
Affiliation(s)
- Joseph G Sinkovics
- The University of South Florida College of Medicine, St. Joseph Hospital's Cancer Institute, Affiliated with the H. L. Moffitt Comprehensive Cancer Center, Tampa, FL 33607-6307, USA.
| |
Collapse
|
15
|
Fowler D, Hoffmann P, Edinger M. Functionally defined T cell subsets in transplantation biology and therapy: regulatory T cells and Th2 cells. Cancer Treat Res 2010; 144:155-86. [PMID: 19779870 DOI: 10.1007/978-0-387-78580-6_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Affiliation(s)
- Daniel Fowler
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | | | | |
Collapse
|
16
|
Ishiyama K, Takami A, Suzuki S, Konaka H, Namiki M, Ooi A, Nakao S. Relationship between tumor-infiltrating T lymphocytes and clinical response after reduced-intensity allogeneic hematopoietic stem cell transplantation for advanced renal cell carcinoma: a single center prospective study. Jpn J Clin Oncol 2009; 39:807-12. [PMID: 19770130 DOI: 10.1093/jjco/hyp104] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE Renal cell carcinoma (RCC) is refractory to conventional therapy, including chemotherapy and radiation. However, because RCC is sensitive to cytokine therapy, an immunotherapeutic approach such as hematopoietic stem cell transplantation (HSCT) might lead to a cure. We performed an institutional clinical study of HSCT for refractory RCC patients. METHODS RCC patients aged 50 years or over, refractory to therapy, were eligible for the study. HSCT was performed after reduced-intensity conditioning. Primary endpoint was defined as the survival at day 100 after HSCT with complete donor chimerism, and secondary endpoint was the effectiveness of HSCT. RESULTS Seven patients, provided with written informed consent, were enrolled in the study. Six of the seven patients achieved complete donor chimera at day 30 after HSCT, but one patient received second HSCT because of graft rejection. Four patients achieved a partial response (PR) and stable disease was observed in another patient, but these responses were temporary. The disease of the other two patients became progressive. Autopsy findings revealed an accumulation of CD8(+) lymphocytes and degenerative changes in the local RCC lesion in three of six patients who responded clinically. An autopsy of a patient who had obtained a PR revealed lymphocyte involvement with a cytotoxic T cell (CTL) phenotype in the metastasis of RCC. CONCLUSIONS Our results demonstrate the efficacy of HSCT for RCC and suggest that the graft-versus-tumor effect elicited by CTLs is induced in vivo. HSCT should be further explored as a potential curative treatment for RCC.
Collapse
Affiliation(s)
- Ken Ishiyama
- Division of Cancer Medicine, Department of Cellular Transplantation Biology, Kanazawa University Graduate School of Medical Science, Kanazawa, Ishikawa, Japan.
| | | | | | | | | | | | | |
Collapse
|