101
|
Bouchlaka MN, Moffitt AB, Kim J, Kink JA, Bloom DD, Love C, Dave S, Hematti P, Capitini CM. Human Mesenchymal Stem Cell-Educated Macrophages Are a Distinct High IL-6-Producing Subset that Confer Protection in Graft-versus-Host-Disease and Radiation Injury Models. Biol Blood Marrow Transplant 2017; 23:897-905. [PMID: 28257800 DOI: 10.1016/j.bbmt.2017.02.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 02/27/2017] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem cells (MSCs) have immunosuppressive and tissue repair properties, but clinical trials using MSCs to prevent or treat graft-versus-host disease (GVHD) have shown mixed results. Macrophages (MØs) are important regulators of immunity and can promote tissue regeneration and remodeling. We have previously shown that MSCs can educate MØs toward a unique anti-inflammatory immunophenotype (MSC-educated MØs [MEMs]); however, their implications for in vivo models of inflammation have not been studied yet. We now show that in comparison with MØs, MEMs have increased expression of the inhibitory molecules PD-L1, PD-L2, in addition to markers of alternatively activated MØs: CD206 and CD163. RNA-Seq analysis of MEMs, as compared with MØs, show a distinct gene expression profile that positively correlates with multiple pathways important in tissue repair. MEMs also show increased expression of IL-6, transforming growth factor-β, arginase-1, CD73, and decreased expression of IL-12 and tumor necrosis factor-α. We show that IL-6 secretion is controlled in part by the cyclo-oxygenase-2, arginase, and JAK1/STAT1 pathway. When tested in vivo, we show that human MEMs significantly enhance survival from lethal GVHD and improve survival of mice from radiation injury. We show these effects could be mediated in part through suppression of human T cell proliferation and may have attenuated host tissue injury in part by enhancing murine fibroblast proliferation. MEMs are a unique MØ subset with therapeutic potential for the management of GVHD and/or protection from radiation-induced injury.
Collapse
Affiliation(s)
- Myriam N Bouchlaka
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Andrea B Moffitt
- Department of Medicine, Duke University, Durham, North Carolina; Duke Center of Genomic and Computational Biology, Durham, North Carolina
| | - Jaehyup Kim
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - John A Kink
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Debra D Bloom
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Cassandra Love
- Department of Medicine, Duke University, Durham, North Carolina; Duke Center of Genomic and Computational Biology, Durham, North Carolina
| | - Sandeep Dave
- Department of Medicine, Duke University, Durham, North Carolina; Duke Center of Genomic and Computational Biology, Durham, North Carolina
| | - Peiman Hematti
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | - Christian M Capitini
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; University of Wisconsin Carbone Cancer Center, Madison, Wisconsin.
| |
Collapse
|
102
|
Godfrey J, Bishop MR, Syed S, Hyjek E, Kline J. PD-1 blockade induces remissions in relapsed classical Hodgkin lymphoma following allogeneic hematopoietic stem cell transplantation. J Immunother Cancer 2017; 5:11. [PMID: 28239465 PMCID: PMC5319147 DOI: 10.1186/s40425-017-0211-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 01/20/2017] [Indexed: 12/24/2022] Open
Abstract
Background Allogeneic hematopoietic stem cell transplantation and checkpoint blockade therapy are immune-based therapies that have activity in selected refractory hematologic malignancies. Interest has developed in combining these treatments for high-risk hematologic diseases. However, there is concern that checkpoint blockade could augment graft-versus-host disease, and very few studies have evaluated the safety of checkpoint blockade in the post-allogeneic setting. Here, we report the outcomes of three patients with relapsed classical Hodgkin’s lymphoma following allogeneic transplant that were treated with the anti-PD-1 antibody, nivolumab. Case presentations Three patients with Hodgkin’s lymphoma relapsed following allogeneic transplant received nivolumab therapy at our institution. All patients were free of graft-versus-host disease and were off of all systemic immunosuppressive medications at the time of nivolumab treatment. Nivolumab was well-tolerated in two of the patients. However, nivolumab had to be discontinued in one patient due to development of immune-related polyarthritis requiring treatment with systemic corticosteroids and methotrexate. Objective responses were observed in all three patients. Conclusions Our case series demonstrates that anti-PD-1 therapy with nivolumab can be highly effective following allogeneic transplant for Hodgkin’s lymphoma, but serious immune-related adverse events can occur, requiring very close monitoring and interruption of therapy.
Collapse
Affiliation(s)
- James Godfrey
- Department of Medicine, University of Chicago, Chicago, IL USA
| | | | - Sahr Syed
- Department Pathology, University of Chicago, Chicago, IL USA
| | - Elizabeth Hyjek
- Department Pathology, University of Chicago, Chicago, IL USA
| | - Justin Kline
- Department of Medicine, University of Chicago, Chicago, IL USA
| |
Collapse
|
103
|
Safety and efficacy of allogeneic hematopoietic stem cell transplant after PD-1 blockade in relapsed/refractory lymphoma. Blood 2017; 129:1380-1388. [PMID: 28073785 DOI: 10.1182/blood-2016-09-738385] [Citation(s) in RCA: 193] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 12/29/2016] [Indexed: 12/24/2022] Open
Abstract
Anti-programmed cell death protein 1 (PD-1) monoclonal antibodies are being increasingly tested in patients with advanced lymphoma. Following treatment, many of those patients are likely to be candidates for allogeneic hematopoietic stem cell transplant (HSCT). However, the safety and efficacy of HSCT may be affected by prior PD-1 blockade. We conducted an international retrospective analysis of 39 patients with lymphoma who received prior treatment with a PD-1 inhibitor, at a median time of 62 days (7-260) before HSCT. After a median follow-up of 12 months, the 1-year cumulative incidences of grade 2-4 and grade 3-4 acute graft-versus-host disease (GVHD) were 44% and 23%, respectively, whereas the 1-year incidence of chronic GVHD was 41%. There were 4 treatment-related deaths (1 from hepatic sinusoidal obstruction syndrome, 3 from early acute GVHD). In addition, 7 patients developed a noninfectious febrile syndrome shortly after transplant requiring prolonged courses of steroids. One-year overall and progression-free survival rates were 89% (95% confidence interval [CI], 74-96) and 76% (95% CI, 56-87), respectively. One-year cumulative incidences of relapse and nonrelapse mortality were 14% (95% CI, 4-29) and 11% (95% CI, 3-23), respectively. Circulating lymphocyte subsets were analyzed in 17 patients. Compared with controls, patients previously treated with PD-1 blockade had significantly decreased PD-1+ T cells and decreased ratios of T-regulatory cells to conventional CD4 and CD8 T cells. In conclusion, HSCT after PD-1 blockade appears feasible with a low rate of relapse. However, there may be an increased risk of early immune toxicity, which could reflect long-lasting immune alterations triggered by prior PD-1 blockade.
Collapse
|
104
|
Tumor regression concomitant with steroid-refractory GvHD highlights the pitfalls of PD-1 blockade following allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant 2017; 52:759-761. [PMID: 28067871 DOI: 10.1038/bmt.2016.346] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
105
|
Jiménez-Ubieto A, Rodriguez A, Martinez Sánchez P, Gómez A, Rodriguez Y, Carreño-Tarragona G, Martinez-López J, Grande C. Fatal graft-versus-host disease after allogeneic stem cell transplantation in a patient recently exposed to nivolumab. J Oncol Pharm Pract 2017; 25:1078155217743069. [PMID: 29207936 DOI: 10.1177/1078155217743069] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation and checkpoint blockade therapy are immune-based salvage therapies for Hodgkin's lymphoma; however, the use of programmed death 1 blocking agents in the allogeneic stem cell transplantation setting could augment the incidence of steroid refractory graft-versus-host disease. Few studies suggest that that nivolumab is safe in patients previously treated with an allogeneic stem cell transplantation. Likewise, there are very limited data on the use of nivolumab before allogeneic stem cell transplantation. Here, we report a case of fatal graft-versus-host disease in a patient who underwent allogeneic stem cell transplantation 26 days after the last administration of nivolumab. Careful monitoring and close clinical assessment of atypical presentation for graft-versus-host disease in these patients, interval of time from nivolumab administration to allogeneic stem cell transplantation, drug dosage adjustments or more effective allo prophilaxys should been evaluated in prospective clinical trial.
Collapse
Affiliation(s)
- Ana Jiménez-Ubieto
- 1 Department of Hematology, 16473 Hospital Universitario 12 de Octubre , Madrid, Spain
| | - Antonia Rodriguez
- 1 Department of Hematology, 16473 Hospital Universitario 12 de Octubre , Madrid, Spain
| | | | - Adolfo Gómez
- 2 Department of Nuclear Medicine, 16473 Hospital Universitario 12 de Octubre , Madrid, Spain
| | - Yolanda Rodriguez
- 3 Department of Pathological Anatomy, 16473 Hospital Universitario 12 de Octubre , Madrid, Spain
| | | | | | - Carlos Grande
- 1 Department of Hematology, 16473 Hospital Universitario 12 de Octubre , Madrid, Spain
| |
Collapse
|
106
|
Hude I, Sasse S, Engert A, Bröckelmann PJ. The emerging role of immune checkpoint inhibition in malignant lymphoma. Haematologica 2017; 102:30-42. [PMID: 27884973 PMCID: PMC5210230 DOI: 10.3324/haematol.2016.150656] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 08/19/2016] [Indexed: 12/19/2022] Open
Abstract
To evade elimination by the host immune system, tumor cells commonly exploit physiological immune checkpoint pathways, restraining efficient anti-tumor immune cell function. Growing understanding of the complex dialog between tumor cells and their microenvironment contributed to the development of immune checkpoint inhibitors. This innovative strategy has demonstrated paradigm-shifting clinical activity in various malignancies. Antibodies targeting programmed death 1 and cytotoxic T-lymphocyte-associated protein-4 are also being investigated in lymphoid malignancies with varying levels of activity and a favorable toxicity profile. To date, evaluated only in the setting of relapsed or refractory disease, anti-programmed death 1 antibodies such as nivolumab and pembrolizumab show encouraging response rates particularly in classical Hodgkin lymphoma but also in follicular lymphoma and diffuse-large B-cell lymphoma. As the first immune checkpoint inhibitor in lymphoma, nivolumab was approved for the treatment of relapsed or refractory classical Hodgkin lymphoma by the Food and Drug Administration in May 2016. In this review, we assess the role of the pathways involved and potential rationale of checkpoint inhibition in various lymphoid malignancies. In addition to data from current clinical trials, immune-related side effects, potential limitations and future perspectives including promising combinatory approaches with immune checkpoint inhibition are discussed.
Collapse
Affiliation(s)
- Ida Hude
- Department of Internal Medicine, Division of Hematology, University Hospital Center Zagreb, Croatia
| | - Stephanie Sasse
- Department I of Internal Medicine and German Hodgkin Study Group (GHSG), University Hospital of Cologne, Germany
| | - Andreas Engert
- Department I of Internal Medicine and German Hodgkin Study Group (GHSG), University Hospital of Cologne, Germany
| | - Paul J Bröckelmann
- Department I of Internal Medicine and German Hodgkin Study Group (GHSG), University Hospital of Cologne, Germany
| |
Collapse
|
107
|
LRBA is Essential for Allogeneic Responses in Bone Marrow Transplantation. Sci Rep 2016; 6:36568. [PMID: 27824136 PMCID: PMC5099895 DOI: 10.1038/srep36568] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 10/18/2016] [Indexed: 12/25/2022] Open
Abstract
The PH-BEACH-WD40 (PBW) protein family members play a role in coordinating receptor signaling and intracellular vesicle trafficking. LPS-Responsive-Beige-like Anchor (LRBA) is a PBW protein whose immune function remains elusive. Here we show that LRBA-null mice are viable, but exhibit compromised rejection of allogeneic, xenogeneic and missing self bone-marrow grafts. Further, we demonstrate that LRBA-null Natural Killer (NK) cells exhibit impaired signaling by the key NK activating receptors, NKp46 and NKG2D. However, induction of IFN-γ by cytokines remains intact, indicating LRBA selectively facilitates signals by receptors for ligands expressed on the surface of NK targets. Surprisingly, LRBA limits immunoregulatory cell numbers in tissues where GvHD is primed or initiated, and consistent with this LRBA-null mice also demonstrate resistance to lethal GvHD. These findings demonstrate that LRBA is redundant for host longevity while being essential for both host and donor-mediated immune responses and thus represents a unique and novel molecular target in transplant immunology.
Collapse
|
108
|
Chronic graft-versus-host disease: biological insights from preclinical and clinical studies. Blood 2016; 129:13-21. [PMID: 27821504 DOI: 10.1182/blood-2016-06-686618] [Citation(s) in RCA: 201] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 07/06/2016] [Indexed: 12/15/2022] Open
Abstract
With the increasing use of mismatched, unrelated, and granulocyte colony-stimulating factor-mobilized peripheral blood stem cell donor grafts and successful treatment of older recipients, chronic graft-versus-host disease (cGVHD) has emerged as the major cause of nonrelapse mortality and morbidity. cGVHD is characterized by lichenoid changes and fibrosis that affects a multitude of tissues, compromising organ function. Beyond steroids, effective treatment options are limited. Thus, new strategies to both prevent and treat disease are urgently required. Over the last 5 years, our understanding of cGVHD pathogenesis and basic biology, born out of a combination of mouse models and correlative clinical studies, has radically improved. We now understand that cGVHD is initiated by naive T cells, differentiating predominantly within highly inflammatory T-helper 17/T-cytotoxic 17 and T-follicular helper paradigms with consequent thymic damage and impaired donor antigen presentation in the periphery. This leads to aberrant T- and B-cell activation and differentiation, which cooperate to generate antibody-secreting cells that cause the deposition of antibodies to polymorphic recipient antigens (ie, alloantibody) or nonpolymorphic antigens common to both recipient and donor (ie, autoantibody). It is now clear that alloantibody can, in concert with colony-stimulating factor 1 (CSF-1)-dependent donor macrophages, induce a transforming growth factor β-high environment locally within target tissue that results in scleroderma and bronchiolitis obliterans, diagnostic features of cGVHD. These findings have yielded a raft of potential new therapeutics, centered on naive T-cell depletion, interleukin-17/21 inhibition, kinase inhibition, regulatory T-cell restoration, and CSF-1 inhibition. This new understanding of cGVHD finally gives hope that effective therapies are imminent for this devastating transplant complication.
Collapse
|
109
|
Robinson TM, Luznik L. For Whom the Bell Tolls: Programmed Death 1 as a Marker of Post-Transplantation Mortality. Biol Blood Marrow Transplant 2016; 22:2115-2116. [PMID: 27638363 DOI: 10.1016/j.bbmt.2016.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 09/08/2016] [Indexed: 10/21/2022]
Affiliation(s)
- Tara M Robinson
- Division of Hematologic Malignancies, Department of Oncology, Johns Hopkins University, Baltimore, Maryland.
| | - Leo Luznik
- Division of Hematologic Malignancies, Department of Oncology, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
110
|
Lin RJ, Diefenbach CS. Checkpoint Inhibition in Hodgkin Lymphoma: Saving the Best for Last? ONCOLOGY (WILLISTON PARK, N.Y.) 2016; 30:914-920. [PMID: 27753058 PMCID: PMC6759920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Hodgkin lymphoma is a unique disease entity characterized by a low number of neoplastic tumor cells surrounded by an inflammatory microenvironment composed of dysfunctional immune cells. Recent molecular and genetic studies have revealed that upregulation of the immune checkpoint pathway programmed death 1/programmed death ligand 1 is a key oncogenic driver of Hodgkin lymphoma. Corroborating these mechanistic studies, early-phase clinical trials using the checkpoint inhibitors nivolumab and pembrolizumab in treatment regimens for relapsed and/or refractory Hodgkin lymphoma have demonstrated impressive response rates, a promising durability of response, and a favorable side-effect profile. Given its targeted mechanism of action, acceptable safety, and clinically meaningful activity, the checkpoint inhibitor nivolumab was recently approved by the US Food and Drug Administration as therapy for classical Hodgkin lymphoma that has relapsed or progressed after autologous stem cell transplantation (ASCT) and post-ASCT consolidation therapy with brentuximab vedotin. In this article we review the scientific rationale, preclinical evidence, and most recent clinical data for the use of checkpoint inhibitor therapy in patients with relapsed Hodgkin lymphoma.
Collapse
|
111
|
Owosho AA, Randazzo J, Rosen EB, Estilo CL, Huryn JM, Chi P, Yom SK. Squamous cell carcinoma associated with chronic graft versus host disease-like/lichen planus-like lesion of the oral cavity in a patient managed for metastatic melanoma with a PD-1 inhibitor pembrolizumab. Oral Oncol 2016; 63:e1-e3. [PMID: 27743938 DOI: 10.1016/j.oraloncology.2016.09.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 09/27/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Adepitan A Owosho
- Dental Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Joseph Randazzo
- Dental Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Evan B Rosen
- Dental Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Cherry L Estilo
- Dental Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Joseph M Huryn
- Dental Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Ping Chi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, United States; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, United States
| | - SaeHee K Yom
- Dental Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.
| |
Collapse
|
112
|
Alatrash G, Daver N, Mittendorf EA. Targeting Immune Checkpoints in Hematologic Malignancies. Pharmacol Rev 2016; 68:1014-1025. [PMID: 27664133 PMCID: PMC11060433 DOI: 10.1124/pr.116.012682] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The use of antibodies that target immune checkpoint molecules on the surface of T-lymphocytes and/or tumor cells has revolutionized our approach to cancer therapy. Cytotoxic-T-lymphocyte antigen (CTLA-4) and programmed cell death protein 1 (PD-1) are the two most commonly targeted immune checkpoint molecules. Although the role of antibodies that target CTLA-4 and PD-1 has been established in solid tumor malignancies and Food and Drug Administration approved for melanoma and non-small cell lung cancer, there remains a desperate need to incorporate immune checkpoint inhibition in hematologic malignancies. Unlike solid tumors, a number of considerations must be addressed to appropriately employ immune checkpoint inhibition in hematologic malignancies. For example, hematologic malignancies frequently obliterate the bone marrow and lymph nodes, which are critical immune organs that must be restored for appropriate response to immune checkpoint inhibition. On the other hand, hematologic malignancies are the quintessential immune responsive tumor type, as proven by the success of allogeneic stem cell transplantation (allo-SCT) in hematologic malignancies. Also, sharing an immune cell lineage, malignant hematologic cells often express immune checkpoint molecules that are absent in solid tumor cells, thereby offering direct targets for immune checkpoint inhibition. A number of clinical trials have demonstrated the potential for immune checkpoint inhibition in hematologic malignancies before and after allo-SCT. The ongoing clinical studies and complimentary immune correlatives are providing a growing body of knowledge regarding the role of immune checkpoint inhibition in hematologic malignancies, which will likely become part of the standard of care for hematologic malignancies.
Collapse
Affiliation(s)
- Gheath Alatrash
- Departments of Stem Cell Transplantation and Cellular Therapy (G.A., E.A.M.), Leukemia (N.D.), and Breast Surgical (E.A.M.) Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naval Daver
- Departments of Stem Cell Transplantation and Cellular Therapy (G.A., E.A.M.), Leukemia (N.D.), and Breast Surgical (E.A.M.) Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elizabeth A Mittendorf
- Departments of Stem Cell Transplantation and Cellular Therapy (G.A., E.A.M.), Leukemia (N.D.), and Breast Surgical (E.A.M.) Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
113
|
Zeiser R, Socié G, Blazar BR. Pathogenesis of acute graft-versus-host disease: from intestinal microbiota alterations to donor T cell activation. Br J Haematol 2016; 175:191-207. [PMID: 27619472 DOI: 10.1111/bjh.14295] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 06/30/2016] [Accepted: 06/30/2016] [Indexed: 01/03/2023]
Abstract
Acute graft-versus-host disease (aGVHD) is a major life-threatening complication of allogeneic haematopoietic cell transplantation (allo-HCT). Here we discuss the aGVHD pathophysiology initiated by multiple signals that cause alloreactive T-cell activation. The outcome of such donor T-cell activation is influenced by T-cell receptor-signal strength, anatomical location, co-stimulatory/co-inhibitory signals and differentiation stage (naive, effector/memory) of T-cells. Additionally, cross-priming of T cells to antigens expressed by pathogens can contribute to aGVHD-mediated tissue injury. In addition to the properties of donor T-cell activation, highly specialized tissue resident cell types, such as innate lymphoid cells, antigen-presenting cells, immune regulatory cells and various intestinal cell populations are critically involved in aGVHD pathogenesis. The role of the thymus and secondary lymphoid tissue injury, non-haematopoietic cells, intestinal microflora, cytokines, chemokines, microRNAs, metabolites and kinases in aGVHD pathophysiology will be highlighted. Acute GVHD pathogenic mechanisms will be connected to novel therapeutic approaches under development for, and tested in, the clinic.
Collapse
Affiliation(s)
- Robert Zeiser
- Department of Haematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Centre, Freiburg, Germany.
| | - Gerard Socié
- Haematology Stem cell transplant Unit, Saint Louis Hospital, APHP, Paris, France
| | - Bruce R Blazar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
114
|
Tsirigotis P, Savani BN, Nagler A. Programmed death-1 immune checkpoint blockade in the treatment of hematological malignancies. Ann Med 2016; 48:428-439. [PMID: 27224873 DOI: 10.1080/07853890.2016.1186827] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The use of tumor-specific monoclonal antibodies (MAbs) has revolutionize the field of cancer immunotherapy. Although treatment of malignant diseases with MAbs is promising, many patients fail to respond or relapse after an initial response. Both solid tumors and hematological malignancies develop mechanisms that enable them to evade the host immune system by usurping immune checkpoint pathways such as PD-1, PD-2, PDL-1, or PDL-2 (programmed cell death protein-1 or 2 and PD-Ligand 1 or 2), which are expressed on activated T cells and on T-regulatory, B cells, natural killers, monocytes, and dendritic cells. One of the most exciting anticancer development in recent years has been the immune checkpoint blockade therapy by using MAbs against immune checkpoint receptor and/or ligands. Anti-PD1 antibodies have been tested in clinical studies that included patients with hematological malignancies and showed remarkable efficacy in Hodgkin lymphoma (HL). In our review, we will focus on the effect of PD-1 activation on hematological malignancies and its role as a therapeutic target. Key messages The programmed death 1 (PD1) immune checkpoint is an important homeostatic mechanism of the immune system that helps in preventing autoimmunity and uncontrolled inflammation in cases of chronic infections. However, PD1 pathway is also operated by a wide variety of malignancies and represents one of the most important mechanisms by which tumor cells escape from the surveillance of the immune system. Blocking of immune checkpoints by the use of monoclonal antibodies opened a new era in the field of cancer immunotherapy. Results from clinical trials are promising, and currently, this approach has been proven effective and safe in patients with solid tumors and hematological malignancies.
Collapse
Affiliation(s)
- Panagiotis Tsirigotis
- a Second Department of Internal Medicine , National and Kapodistrian University of Athens , Athens , Greece
| | - Bipin N Savani
- b Department of Hematology, Vanderbilt University Medical Center , Nashville , TN , USA
| | - Arnon Nagler
- c Hematology Division , Chaim Sheba Medical Center , Tel Hashomer , Israel
| |
Collapse
|
115
|
Singh AK, Porrata LF, Aljitawi O, Lin T, Shune L, Ganguly S, McGuirk JP, Abhyankar S. Fatal GvHD induced by PD-1 inhibitor pembrolizumab in a patient with Hodgkin's lymphoma. Bone Marrow Transplant 2016; 51:1268-1270. [PMID: 27111048 DOI: 10.1038/bmt.2016.111] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- A K Singh
- Division of Hematologic Malignancies and Cellular Therapeutics, Blood and Marrow Transplantation Program, University of Kansas Medical Center, Westwood, KS, USA
| | - L F Porrata
- Department of Hematology, Mayo Clinic, Rochester, MN, USA
| | - O Aljitawi
- Division of Hematologic Malignancies and Cellular Therapeutics, Blood and Marrow Transplantation Program, University of Kansas Medical Center, Westwood, KS, USA
| | - T Lin
- Division of Hematologic Malignancies and Cellular Therapeutics, Blood and Marrow Transplantation Program, University of Kansas Medical Center, Westwood, KS, USA
| | - L Shune
- Division of Hematologic Malignancies and Cellular Therapeutics, Blood and Marrow Transplantation Program, University of Kansas Medical Center, Westwood, KS, USA
| | - S Ganguly
- Division of Hematologic Malignancies and Cellular Therapeutics, Blood and Marrow Transplantation Program, University of Kansas Medical Center, Westwood, KS, USA
| | - J P McGuirk
- Division of Hematologic Malignancies and Cellular Therapeutics, Blood and Marrow Transplantation Program, University of Kansas Medical Center, Westwood, KS, USA
| | - S Abhyankar
- Division of Hematologic Malignancies and Cellular Therapeutics, Blood and Marrow Transplantation Program, University of Kansas Medical Center, Westwood, KS, USA
| |
Collapse
|
116
|
Herbel C, Patsoukis N, Bardhan K, Seth P, Weaver JD, Boussiotis VA. Clinical significance of T cell metabolic reprogramming in cancer. Clin Transl Med 2016; 5:29. [PMID: 27510264 PMCID: PMC4980327 DOI: 10.1186/s40169-016-0110-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 07/15/2016] [Indexed: 02/06/2023] Open
Abstract
Conversion of normal cells to cancer is accompanied with changes in their metabolism. During this conversion, cell metabolism undergoes a shift from oxidative phosphorylation to aerobic glycolysis, also known as Warburg effect, which is a hallmark for cancer cell metabolism. In cancer cells, glycolysis functions in parallel with the TCA cycle and other metabolic pathways to enhance biosynthetic processes and thus support proliferation and growth. Similar metabolic features are observed in T cells during activation but, in contrast to cancer, metabolic transitions in T cells are part of a physiological process. Currently, there is intense interest in understanding the cause and effect relationship between metabolic reprogramming and T cell differentiation. After the recent success of cancer immunotherapy, the crosstalk between immune system and cancer has come to the forefront of clinical and basic research. One of the key goals is to delineate how metabolic alterations of cancer influence metabolism-regulated function and differentiation of tumor resident T cells and how such effects might be altered by immunotherapy. Here, we review the unique metabolic features of cancer, the implications of cancer metabolism on T cell metabolic reprogramming during antigen encounters, and the translational prospective of harnessing metabolism in cancer and T cells for cancer therapy.
Collapse
Affiliation(s)
- Christoph Herbel
- Division of Hematology-Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Nikolaos Patsoukis
- Division of Hematology-Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Kankana Bardhan
- Division of Hematology-Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Pankaj Seth
- Division of Hematology-Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA.,Beth Israel Deaconess Cancer Center, Harvard Medical School, 330 Brookline Avenue, Dana 513, Boston, MA, 02215, USA.,Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical Center, Boston, USA
| | - Jessica D Weaver
- Division of Hematology-Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Vassiliki A Boussiotis
- Division of Hematology-Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA. .,Beth Israel Deaconess Cancer Center, Harvard Medical School, 330 Brookline Avenue, Dana 513, Boston, MA, 02215, USA.
| |
Collapse
|
117
|
Schade H, Sen S, Neff CP, Freed BM, Gao D, Gutman JA, Palmer BE. Programmed Death 1 Expression on CD4 + T Cells Predicts Mortality after Allogeneic Stem Cell Transplantation. Biol Blood Marrow Transplant 2016; 22:2172-2179. [PMID: 27519280 DOI: 10.1016/j.bbmt.2016.08.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 08/08/2016] [Indexed: 12/22/2022]
Abstract
Excessive or persistent programmed death 1 (PD-1) expression on virus- or tumor-specific T cells during chronic viral infection or malignancy has been associated with impaired immune control. To assess the role of the PD-1 pathway in allogeneic stem cell transplantation (SCT), we examined PD-1 expression and maturation phenotype on T cells from 42 patients early (day 55 to 85) after cord blood (CB), matched unrelated donor, and matched related donor transplantation. Expression of PD-1 on CD4+ T cells was significantly elevated in all transplantation types, with the highest level observed in CB subjects. Elevated PD-1 expression on CD4+ T cells early after transplantation was observed in nonsurvivors (median, 40.2%; range, 15.1 to 86.1) compared with survivors (median, 23.6%; range, 8.4 to 55.2; P = .001), indicating its association with increased risk for mortality, especially with CB transplantations, where PD-1 was increased in nonsurvivors (median, 64.6%; range, 36.5 to 86.1) compared with survivors (median, 34.1%; range, 15.9 to 55.2; P = .01). Furthermore, T cell subset analysis revealed that PD-1 expression was further elevated on CD4+ T central memory in nonsurvivors (median, 49.8%; range, 15.1 to 83.4) compared with survivors (median, 24.8%; range, 8.9 to 71.3; P = .002) and on T effector memory cells in nonsurvivors (median, 69.1%; range, 24.7 to 92.6) compared with survivors (median, 43.7%; range, 13.9 to 96.5; P = .0003). Our findings suggest that elevation of PD-1 expression on CD4+ T cells is associated with mortality in CB and possibly all SCT recipients.
Collapse
Affiliation(s)
- Henning Schade
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Division of Hematology and Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sharon Sen
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Division of Allergy and Clinical Immunology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - C Preston Neff
- Division of Allergy and Clinical Immunology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Brian M Freed
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Division of Allergy and Clinical Immunology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Dexiang Gao
- Department of Pediatrics and Department of Biostatistics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jonathan A Gutman
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Division of Hematology and Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Brent E Palmer
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Division of Allergy and Clinical Immunology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
118
|
Alterations of circulating lymphoid committed progenitor cellular metabolism after allogeneic stem cell transplantation in humans. Exp Hematol 2016; 44:811-816.e3. [PMID: 27321893 DOI: 10.1016/j.exphem.2016.05.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 05/13/2016] [Accepted: 05/17/2016] [Indexed: 02/01/2023]
Abstract
Lymphoid-committed CD34(+)lin(-)CD10(+)CD24(-) progenitors undergo a rebound at month 3 after allogeneic hematopoietic stem cell transplantation (allo-HSCT) in the absence of acute graft-versus-host disease (aGVHD). Here, we analyzed transcriptional programs of cell-sorted circulating lymphoid-committed progenitors and CD34(+)Lin(-)CD10(-) nonlymphoid progenitors in 11 allo-HSCT patients who had (n = 5) or had not (n = 6) developed grade 2 or 3 aGVHD and in 7 age-matched healthy donors. Major upregulated pathways include protein synthesis, energy production, cell cycle regulation, and cytoskeleton organization. Notably, genes from protein biogenesis, translation machinery, and cell cycle (CDK6) were overexpressed in progenitors from patients in the absence of aGVHD compared with healthy donors and patients affected by aGVHD. Expression of many genes from the mitochondrial oxidative phosphorylation metabolic pathway leading to ATP production were more specifically increased in lymphoid-committed progenitors in the absence of aGVHD. This was also the case for genes involved in cell mobilization such as those regulating Rho GTPase activity. In all, we found that circulating lymphoid-committed progenitors undergo profound changes in metabolism, favoring cell proliferation, energy production, and cell mobilization after allo-HSCT in humans. These mechanisms are abolished in the case of aGVHD or its treatment, indicating a persistent cell-intrinsic defect after exit from the bone marrow.
Collapse
|
119
|
Saha A, O'Connor RS, Thangavelu G, Lovitch SB, Dandamudi DB, Wilson CB, Vincent BG, Tkachev V, Pawlicki JM, Furlan SN, Kean LS, Aoyama K, Taylor PA, Panoskaltsis-Mortari A, Foncea R, Ranganathan P, Devine SM, Burrill JS, Guo L, Sacristan C, Snyder NW, Blair IA, Milone MC, Dustin ML, Riley JL, Bernlohr DA, Murphy WJ, Fife BT, Munn DH, Miller JS, Serody JS, Freeman GJ, Sharpe AH, Turka LA, Blazar BR. Programmed death ligand-1 expression on donor T cells drives graft-versus-host disease lethality. J Clin Invest 2016; 126:2642-60. [PMID: 27294527 DOI: 10.1172/jci85796] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 04/28/2016] [Indexed: 12/20/2022] Open
Abstract
Programmed death ligand-1 (PD-L1) interaction with PD-1 induces T cell exhaustion and is a therapeutic target to enhance immune responses against cancer and chronic infections. In murine bone marrow transplant models, PD-L1 expression on host target tissues reduces the incidence of graft-versus-host disease (GVHD). PD-L1 is also expressed on T cells; however, it is unclear whether PD-L1 on this population influences immune function. Here, we examined the effects of PD-L1 modulation of T cell function in GVHD. In patients with severe GVHD, PD-L1 expression was increased on donor T cells. Compared with mice that received WT T cells, GVHD was reduced in animals that received T cells from Pdl1-/- donors. PD-L1-deficient T cells had reduced expression of gut homing receptors, diminished production of inflammatory cytokines, and enhanced rates of apoptosis. Moreover, multiple bioenergetic pathways, including aerobic glycolysis, oxidative phosphorylation, and fatty acid metabolism, were also reduced in T cells lacking PD-L1. Finally, the reduction of acute GVHD lethality in mice that received Pdl1-/- donor cells did not affect graft-versus-leukemia responses. These data demonstrate that PD-L1 selectively enhances T cell-mediated immune responses, suggesting a context-dependent function of the PD-1/PD-L1 axis, and suggest selective inhibition of PD-L1 on donor T cells as a potential strategy to prevent or ameliorate GVHD.
Collapse
|
120
|
Kouidhi S, Noman MZ, Kieda C, Elgaaied AB, Chouaib S. Intrinsic and Tumor Microenvironment-Induced Metabolism Adaptations of T Cells and Impact on Their Differentiation and Function. Front Immunol 2016; 7:114. [PMID: 27066006 PMCID: PMC4810024 DOI: 10.3389/fimmu.2016.00114] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 03/14/2016] [Indexed: 01/09/2023] Open
Abstract
It is well recognized that the immune system and metabolism are highly integrated. In this context, multilevel interactions between metabolic system and T lymphocyte signaling and fate exist. This review will discuss different potential cell metabolism pathways involved in shaping T lymphocyte function and differentiation. We will also provide a general framework for understanding how tumor microenvironmental metabolism, associated with hypoxic stress, interferes with T-cell priming and expansion. How T-cell metabolism drives T-cell-mediated immunity and how the manipulation of metabolic programing for therapeutic purposes will be also discussed.
Collapse
Affiliation(s)
- Soumaya Kouidhi
- Laboratory BVBGR, LR11ES31, ISBST, Higher Institute of Biotechnology of Sidi Thabet, University of Manouba, Tunis, Tunisia; Laboratory of Genetics, Immunology and Human Pathology, Faculty of Sciences of Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Muhammad Zaeem Noman
- Laboratory «Integrative Tumor Immunology and Genetic Oncology» Equipe Labellisée LIGUE 2015, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1186, Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Gustave Roussy, Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Claudine Kieda
- Centre de Biophysique Moléculaire, CNRS UPR 4301 , Orléans , France
| | - Amel Benammar Elgaaied
- Laboratory of Genetics, Immunology and Human Pathology, Faculty of Sciences of Tunis, University Tunis El Manar , Tunis , Tunisia
| | - Salem Chouaib
- Laboratory «Integrative Tumor Immunology and Genetic Oncology» Equipe Labellisée LIGUE 2015, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1186, Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Gustave Roussy, Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
121
|
Magenau J, Runaas L, Reddy P. Advances in understanding the pathogenesis of graft-versus-host disease. Br J Haematol 2016; 173:190-205. [PMID: 27019012 DOI: 10.1111/bjh.13959] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 12/15/2015] [Indexed: 01/24/2023]
Abstract
Allogeneic haematopoietic stem cell transplantation (HCT) is a potent immunotherapy with curative potential for several haematological disorders. Overcoming the immunological barrier of acute graft-versus-host disease (GVHD) remains a fundamental impediment to expanding the efficacy of HCT. GVHD reflects a complex pathological interaction between the innate and adaptive immune systems of the host and donor. Over the past decade there has been a tremendous advancement in our understanding of the cellular and molecular underpinnings of this devastating disease. In this review, we cover several recently appreciated facets of GVHD pathogenesis including novel extracellular mediators of inflammation, immune subsets, intracellular signal transduction, post-translation modifications and epigenetic regulation. We begin to develop general themes regarding the immunological pathways in GVHD pathogenesis, discuss critical outstanding questions, and explore new avenues for GVHD treatment and prevention.
Collapse
Affiliation(s)
- John Magenau
- Blood and Marrow Transplant Program, Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Lyndsey Runaas
- Blood and Marrow Transplant Program, Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Pavan Reddy
- Blood and Marrow Transplant Program, Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
122
|
Villa NY, Rahman MM, McFadden G, Cogle CR. Therapeutics for Graft-versus-Host Disease: From Conventional Therapies to Novel Virotherapeutic Strategies. Viruses 2016; 8:85. [PMID: 27011200 PMCID: PMC4810275 DOI: 10.3390/v8030085] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 03/09/2016] [Accepted: 03/09/2016] [Indexed: 02/06/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) has a curative potential for many hematologic malignancies and blood diseases. However, the success of allo-HSCT is limited by graft-versus-host disease (GVHD), an immunological syndrome that involves inflammation and tissue damage mediated by donor lymphocytes. Despite immune suppression, GVHD is highly incident even after allo-HSCT using human leukocyte antigen (HLA)-matched donors. Therefore, alternative and more effective therapies are needed to prevent or control GVHD while preserving the beneficial graft-versus-cancer (GVC) effects against residual disease. Among novel therapeutics for GVHD, oncolytic viruses such as myxoma virus (MYXV) are receiving increased attention due to their dual role in controlling GVHD while preserving or augmenting GVC. This review focuses on the molecular basis of GVHD, as well as state-of-the-art advances in developing novel therapies to prevent or control GVHD while minimizing impact on GVC. Recent literature regarding conventional and the emerging therapies are summarized, with special emphasis on virotherapy to prevent GVHD. Recent advances using preclinical models with oncolytic viruses such as MYXV to ameliorate the deleterious consequences of GVHD, while maintaining or improving the anti-cancer benefits of GVC will be reviewed.
Collapse
Affiliation(s)
- Nancy Y Villa
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL 32610, USA.
| | - Masmudur M Rahman
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA.
| | - Grant McFadden
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA.
| | - Christopher R Cogle
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
123
|
Perales MA, Sauter CS, Armand P. Reprint of: Fast Cars and No Brakes: Autologous Stem Cell Transplantation as a Platform for Novel Immunotherapies. Biol Blood Marrow Transplant 2016; 22:S9-S14. [PMID: 26899275 DOI: 10.1016/j.bbmt.2016.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 10/14/2015] [Indexed: 11/24/2022]
Abstract
Autologous stem cell transplantation (ASCT) is indicated in a number of hematologic malignancies, including multiple myeloma, non-Hodgkin lymphoma, and Hodgkin lymphoma. Relapse, however, remains 1 of the main causes of post-ASCT failure, and several strategies are being investigated to decrease the risk of relapse of progression. Recent advances in the treatment of hematological malignancies have included adoptive transfer of genetically modified T cells that express chimeric antigen receptors or T cell receptors, as well the use of checkpoint inhibitors. Early clinical results in non-transplantation patients have been very promising. This review will focus on the use of gene-modified T cells and checkpoint inhibitors in stem cell transplantation.
Collapse
Affiliation(s)
- Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York.
| | - Craig S Sauter
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Philippe Armand
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
124
|
Engelhardt BG, Paczesny S, Jung DK, Daguindau E, Jagasia M, Savani BN, Chinratanalab W, Cornell RF, Goodman S, Greer JP, Kassim AA, Sengsayadeth S, Yoder SM, Rock MT, Crowe JE. Early Th1 immunity promotes immune tolerance and may impair graft-versus-leukemia effect after allogeneic hematopoietic cell transplantation. Haematologica 2016; 101:e204-8. [PMID: 26819055 DOI: 10.3324/haematol.2015.139501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Brian G Engelhardt
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sophie Paczesny
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA Department of Microbiology and Immunology, Indiana University School, Indianapolis, IN, USA
| | - Dae Kwang Jung
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Etienne Daguindau
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA Department of Microbiology and Immunology, Indiana University School, Indianapolis, IN, USA
| | - Madan Jagasia
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bipin N Savani
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wichai Chinratanalab
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Robert F Cornell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Stacey Goodman
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John P Greer
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Adetola A Kassim
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Salyka Sengsayadeth
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sandra M Yoder
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael T Rock
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James E Crowe
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA Department of Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
125
|
Bruno B, Auner HW, Gahrton G, Garderet L, Festuccia M, Ladetto M, Lemoli RM, Massaia M, Morris C, Palumbo A, Schönland S, Boccadoro M, Kröger N. Stem cell transplantation in multiple myeloma and other plasma cell disorders (report from an EBMT preceptorship meeting). Leuk Lymphoma 2016; 57:1256-68. [PMID: 26735310 DOI: 10.3109/10428194.2015.1131278] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The European Society for Blood and Marrow Transplantation Chronic Malignancies Working Party held a preceptorship meeting in Turin, Italy on 25-26 September 2014, to discuss the role of stem cell transplantation (SCT) in the treatment of multiple myeloma and other plasma cell disorders. Scientists and clinicians working in the field gathered to discuss a variety of topics including the results of recent clinical trials, basic research, the concept of minimal residual disease, and immune modulation. As individual presentations revealed, important advances have occurred in our understanding of the pathophysiology of myeloma and the role that SCT, along with other forms of immunotherapy, plays in treating it. Each presentation stimulated discussion and exchange of ideas among the attendants. We decided to summarize and, importantly, to update the meeting proceedings in this review to share stimulating discussions and ideas on potentially novel treatment strategies among clinicians.
Collapse
Affiliation(s)
- Benedetto Bruno
- a Division of Hematology , University of Torino , Torino , Italy ;,b Department of Molecular Biotechnology and Health Sciences , University of Torino , Torino , Italy
| | - Holger W Auner
- c Centre for Hematology, Imperial College London , London , UK
| | - Gösta Gahrton
- d Department of Medicine , Karolinska Institutet , Stockholm , Sweden
| | - Laurent Garderet
- e INSERM, UMR_S 938, Proliferation and Differentiation of Stem Cells , Paris , France
| | - Moreno Festuccia
- a Division of Hematology , University of Torino , Torino , Italy ;,b Department of Molecular Biotechnology and Health Sciences , University of Torino , Torino , Italy
| | - Marco Ladetto
- f Division of Hematology, Azienda Ospedaliera SS Antonio E Biagio E Cesare Arrigo , Alessandria , Italy
| | - Roberto M Lemoli
- g AOU IRCCS S. Martino-IST, Università Di Genova , Genova, Italy
| | - Massimo Massaia
- h Division of Hematology and Cell Therapy, Ordine Mauriziano Hospital, Department of Molecular Biotechnology and Health Sciences , University of Torino , Torino , Italy
| | - Curly Morris
- i Centre for Cancer Research and Cell Biology , Queen's University of Belfast , Belfast , UK
| | - Antonio Palumbo
- a Division of Hematology , University of Torino , Torino , Italy ;,b Department of Molecular Biotechnology and Health Sciences , University of Torino , Torino , Italy
| | - Stefan Schönland
- j Medical Department V , Amyloidosis Center , Heidelberg , Germany
| | - Mario Boccadoro
- a Division of Hematology , University of Torino , Torino , Italy ;,b Department of Molecular Biotechnology and Health Sciences , University of Torino , Torino , Italy
| | - Nicolaus Kröger
- k Department of Stem Cell Transplantation , University Hospital Hamburg , Hamburg , Germany
| |
Collapse
|
126
|
Michonneau D, Sagoo P, Breart B, Garcia Z, Celli S, Bousso P. The PD-1 Axis Enforces an Anatomical Segregation of CTL Activity that Creates Tumor Niches after Allogeneic Hematopoietic Stem Cell Transplantation. Immunity 2016; 44:143-154. [DOI: 10.1016/j.immuni.2015.12.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 09/18/2015] [Accepted: 10/12/2015] [Indexed: 12/21/2022]
|
127
|
Jezeršek Novaković B. Checkpoint inhibitors in Hodgkin's lymphoma. Eur J Haematol 2015; 96:335-43. [PMID: 26560962 DOI: 10.1111/ejh.12697] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2015] [Indexed: 12/11/2022]
Abstract
Hodgkin's lymphoma is unusual among cancers in that it consists of a small number of malignant Hodgkin/Reed-Sternberg cells in a sea of immune system cells, including T cells. Most of these T cells are reversibly inactivated in different ways and their reactivation may induce a very strong immune response to cancer cells. One way of reactivation of T cells is with antibodies blocking the CTLA-4 and especially with antibodies directed against PD-1 or the PD-L1 ligand thereby reversing the tumor-induced downregulation of T-cell function and augmenting antitumor immune activity at the priming (CTLA-4) or tissue effector (PD-1) phase. Immune checkpoint inhibitors have been evidenced as an additional treatment option with substantial effectiveness and acceptable toxicity in heavily pretreated patients with Hodgkin's lymphoma. Particularly, PD-1 blockade with nivolumab and pembrolizumab has demonstrated significant single-agent activity in this select population.
Collapse
|
128
|
Nivolumab in a patient with refractory Hodgkin's lymphoma after allogeneic stem cell transplantation. Bone Marrow Transplant 2015; 51:443-5. [PMID: 26551782 DOI: 10.1038/bmt.2015.266] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
129
|
Fast Cars and No Brakes: Autologous Stem Cell Transplantation as a Platform for Novel Immunotherapies. Biol Blood Marrow Transplant 2015; 22:17-22. [PMID: 26485445 DOI: 10.1016/j.bbmt.2015.10.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 10/14/2015] [Indexed: 01/21/2023]
Abstract
Autologous stem cell transplantation (ASCT) is indicated in a number of hematologic malignancies, including multiple myeloma, non-Hodgkin lymphoma, and Hodgkin lymphoma. Relapse, however, remains 1 of the main causes of post-ASCT failure, and several strategies are being investigated to decrease the risk of relapse of progression. Recent advances in the treatment of hematological malignancies have included adoptive transfer of genetically modified T cells that express chimeric antigen receptors or T cell receptors, as well the use of checkpoint inhibitors. Early clinical results in nontransplantation patients have been very promising. This review will focus on the use of gene-modified T cells and checkpoint inhibitors in stem cell transplantation.
Collapse
|
130
|
Slack M, Wang T, Wang R. T cell metabolic reprogramming and plasticity. Mol Immunol 2015; 68:507-12. [PMID: 26277274 DOI: 10.1016/j.molimm.2015.07.036] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 07/26/2015] [Indexed: 12/21/2022]
Abstract
Upon antigen stimulation, small and quiescent naïve T cells undergo an approximately 24h growth phase followed by rapid proliferation. Depending on the nature of the antigen and cytokine milieu, these proliferating T cells differentiate into distinctive functional subgroups that are essential for appropriate immune defense and regulation. T cells undergo a characteristic metabolic rewiring that fulfills the dramatically increased bioenergetic and biosynthetic demands during the transition between resting, activation and differentiation. Beyond this, T cells are distributed throughout the body and are able to function in a wide range of physio-pathological environments, including some with a dramatic metabolic derangement. As such, T cells must quickly respond to and adapt to fluctuations in environmental nutrient levels. We consider such responsiveness and adaptation in terms of metabolic plasticity, that is, an evolutionarilly selected process which allows T cells to illicit robust immune functions in response to either a continuous or disrupted nutrient supply. In this review, we illustrate the relevant metabolic pathways in T cells and discuss the ability of T cells to change their metabolic substrates in response to changes in the environment.
Collapse
Affiliation(s)
- Maria Slack
- Department of Pediatrics, The Ohio State University School of Medicine, Columbus, OH, USA; Division of Allergy and Immunology Nationwide Children's Hospital, Columbus, OH, USA; Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, OH, USA
| | - Tingting Wang
- Center for Childhood Cancer and Blood Disease, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Ruoning Wang
- Center for Childhood Cancer and Blood Disease, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Hematology/Oncology & BMT, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University School of Medicine, Columbus, OH, USA.
| |
Collapse
|
131
|
Tkachev V, Goodell S, Opipari AW, Hao LY, Franchi L, Glick GD, Ferrara JLM, Byersdorfer CA. Programmed death-1 controls T cell survival by regulating oxidative metabolism. THE JOURNAL OF IMMUNOLOGY 2015; 194:5789-800. [PMID: 25972478 DOI: 10.4049/jimmunol.1402180] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 04/14/2015] [Indexed: 12/13/2022]
Abstract
The coinhibitory receptor programmed death-1 (PD-1) maintains immune homeostasis by negatively regulating T cell function and survival. Blockade of PD-1 increases the severity of graft-versus-host disease (GVHD), but the interplay between PD-1 inhibition and T cell metabolism is not well studied. We found that both murine and human alloreactive T cells concomitantly upregulated PD-1 expression and increased levels of reactive oxygen species (ROS) following allogeneic bone marrow transplantation. This PD-1(Hi)ROS(Hi) phenotype was specific to alloreactive T cells and was not observed in syngeneic T cells during homeostatic proliferation. Blockade of PD-1 signaling decreased both mitochondrial H2O2 and total cellular ROS levels, and PD-1-driven increases in ROS were dependent upon the oxidation of fatty acids, because treatment with etomoxir nullified changes in ROS levels following PD-1 blockade. Downstream of PD-1, elevated ROS levels impaired T cell survival in a process reversed by antioxidants. Furthermore, PD-1-driven changes in ROS were fundamental to establishing a cell's susceptibility to subsequent metabolic inhibition, because blockade of PD-1 decreased the efficacy of later F1F0-ATP synthase modulation. These data indicate that PD-1 facilitates apoptosis in alloreactive T cells by increasing ROS in a process dependent upon the oxidation of fat. In addition, blockade of PD-1 undermines the potential for subsequent metabolic inhibition, an important consideration given the increasing use of anti-PD-1 therapies in the clinic.
Collapse
Affiliation(s)
- Victor Tkachev
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109
| | - Stefanie Goodell
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109
| | - Anthony W Opipari
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109
| | | | | | - Gary D Glick
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109; and
| | - James L M Ferrara
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109
| | - Craig A Byersdorfer
- Division of Blood and Marrow Transplant and Cellular Therapies, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224
| |
Collapse
|
132
|
Abstract
Programmed cell death 1 (PD-1) is an inducible immune modulatory receptor. Upon interaction with its ligands B7 homolog 1 (PD-L1) and B7-DC (PD-L2), PD-1 plays important roles in negative regulation of T cell responses to antigen stimulation and maintaining peripheral tolerance. In addition to the inducible expression pattern on conventional T cells, PD-1 is also found on regulatory T cells, follicular T and B cells, and antigen-presenting cells including activated dendritic cells and monocytes. Therefore, PD-1 may have a much broader functionality than expected in negative regulation of multiple arms of immune responses. In addition to cancer therapy, the manipulation of PD-1 and its ligands may hold great promise for therapeutic applications also in autoimmune and infectious diseases.
Collapse
|
133
|
Festuccia M, Martino M, Ferrando F, Messina G, Moscato T, Fedele R, Boccadoro M, Giaccone L, Bruno B. Allogeneic stem cell transplantation in multiple myeloma: immunotherapy and new drugs. Expert Opin Biol Ther 2015; 15:857-72. [PMID: 25865214 DOI: 10.1517/14712598.2015.1036735] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Autologous (auto) stem cell transplantation (SCT) and the development of new drugs have improved the survival of multiple myeloma (MM) patients. By contrast, though potentially curative, the use of allogeneic (allo)-SCT is controversial. AREAS COVERED A review has been conducted to examine the current evidence for the use of allo-SCT in MM. We have examined novel cell therapies that may be exploited to induce myeloma-specific immune responses including the new promising frontier of chimeric antigen receptor (CAR)-T and -natural killer (NK) cells. EXPERT OPINION One of the major controversies facing researchers in exploring the allo approach is the remarkable recent treatment improvement observed with second- and third-generation proteasome inhibitors and immunomodulatory drugs, monoclonal antibodies and deacetylase inhibitors. Despite these great advances, the disease remains to be incurable and allo-SCT may still play a role in the cure of MM. We think that allo-SCT conserves a role in MM and its curative potential in high-risk patients should be explored in the setting of control clinical trials. Novel cell therapies such as CAR technologies may open new avenues of research toward a potential cure. Data from currently ongoing prospective studies will be helpful to clarify pending clinical questions.
Collapse
Affiliation(s)
- Moreno Festuccia
- University of Torino, Presidio Molinette, and Department of Molecular Biotechnology and Health Sciences, Division of Hematology, A.O.U. Citta' della salute e della scienza di Torino, Presidio Molinette , Torino , Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Immune checkpoint blockade in hematologic malignancies. Blood 2015; 125:3393-400. [PMID: 25833961 DOI: 10.1182/blood-2015-02-567453] [Citation(s) in RCA: 185] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 03/25/2015] [Indexed: 02/07/2023] Open
Abstract
Therapeutic blockade of immune checkpoint pathways, in particular cytotoxic T-lymphocyte associated protein 4 and programmed-death 1 (PD-1), has become a paradigm-shifting treatment in solid tumor oncology. Hematologic malignancies (HMs), many of which are known to have clinically exploitable immune sensitivity, are a natural target for this type of treatment. Several clinical trials of checkpoint blockade have been conducted in HM, with preliminary results suggesting the therapeutic usefulness of this approach across several tumor types. In particular, the results of PD-1 blockade in Hodgkin lymphoma (HL) are remarkable, and raise hope that it may alter the treatment landscape in this disease. However, numerous questions remain about the optimal role of checkpoint blockade both in HL and beyond. Those questions are the focus of this review, in the hope that, if we are at the dawn of a new day in HM immunotherapy, we may begin to envision its morning.
Collapse
|
135
|
B7-H3 expression in donor T cells and host cells negatively regulates acute graft-versus-host disease lethality. Blood 2015; 125:3335-46. [PMID: 25814530 DOI: 10.1182/blood-2014-09-603357] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 03/10/2015] [Indexed: 12/18/2022] Open
Abstract
Members of the B7 family have been shown to be important for regulating immune responses by providing either positive or negative costimulatory signals. The function of B7-H3 has been controversial. We show that B7-H3 is upregulated in graft-versus-host disease (GVHD) target organs, including the colon, liver, and lung. Infusion of allogeneic donor T cells into B7-H3(-/-) vs wild-type (WT) recipients resulted in increased GVHD lethality associated with increased T-cell proliferation, colonic inflammatory cytokines, and destruction of epithelial barriers. Allogeneic B7-H3(-/-) vs WT donor T cells also had increased T-cell proliferation and GVHD lethality associated with increased proliferation and cytokine secretion in the spleen, intraepithelial lymphocyte inflammatory cytokines, and intestinal permeability. Both resting and activated regulatory T cells (Tregs) lack B7-H3 messenger RNA. Consistent with these data, GVHD was augmented in recipients of B7-H3(-/-) Treg-depleted grafts. In two delayed lymphocyte infusion (DLI) models, T cells lacking B7-H3 are capable of providing graft-versus-leukemia (GVL) effects. We conclude that B7-H3 is responsible for providing a negative costimulatory signal. Our studies provide support for developing and testing new therapies directed toward the B7-H3 pathway, including approaches to augment host B7-H3 early after bone marrow transplantation to prevent GVHD and to develop potent antagonistic antibodies later after transplant to facilitate DLI-mediated GVL without GVHD complications.
Collapse
|
136
|
BET bromodomain inhibition suppresses graft-versus-host disease after allogeneic bone marrow transplantation in mice. Blood 2015; 125:2724-8. [PMID: 25778533 DOI: 10.1182/blood-2014-08-598037] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 03/05/2015] [Indexed: 02/06/2023] Open
Abstract
Acute graft-versus-host disease (GVHD) is the major obstacle of allogeneic bone marrow transplantation (BMT). Bromodomain and extra-terminal (BET) protein inhibitors selectively block acetyl-binding pockets of the bromodomains and modulate histone acetylation. Here, we report that inhibition of BET bromodomain (BRD) proteins with I-BET151 alters cytokine expression in dendritic cells (DCs) and T cells, including surface costimulatory molecules, in vitro and in vivo cytokine secretion, and expansion. Mechanistic studies with I-BET151 and JQ1, another inhibitor, demonstrate that these effects could be from disruption of association between BRD4 and acetyl-310 RelA of nuclear factor kappa B. Short-term administration early during BMT reduced GVHD severity and improved mortality in two different allogeneic BMT models but retained sufficient graft-versus-tumor effect. Thus inhibiting BRD proteins may serve as a novel approach for preventing GVHD.
Collapse
|
137
|
T cell metabolic fitness in antitumor immunity. Trends Immunol 2015; 36:257-64. [PMID: 25773310 DOI: 10.1016/j.it.2015.02.007] [Citation(s) in RCA: 232] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 02/17/2015] [Accepted: 02/18/2015] [Indexed: 01/08/2023]
Abstract
T cell metabolism has a central role in supporting and shaping immune responses and may have a key role in antitumor immunity. T cell metabolism is normally held under tight regulation in an immune response of glycolysis to promote effector T cell expansion and function. However, tumors may deplete nutrients, generate toxic products, or stimulate conserved negative feedback mechanisms, such as through Programmed Cell Death 1 (PD-1), to impair effector T cell nutrient uptake and metabolic fitness. In addition, regulatory T cells are favored in low glucose conditions and may inhibit antitumor immune responses. Here, we review how the tumor microenvironment modifies metabolic and functional pathways in T cells and how these changes may uncover new targets and challenges for cancer immunotherapy and treatment.
Collapse
|
138
|
Ritprajak P, Azuma M. Intrinsic and extrinsic control of expression of the immunoregulatory molecule PD-L1 in epithelial cells and squamous cell carcinoma. Oral Oncol 2014; 51:221-8. [PMID: 25500094 DOI: 10.1016/j.oraloncology.2014.11.014] [Citation(s) in RCA: 261] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Revised: 11/13/2014] [Accepted: 11/20/2014] [Indexed: 01/01/2023]
Abstract
Recent clinical results for PD-1 blockade therapy have demonstrated durable tumor control with minimal immune-related adverse effects. PD-L1 is induced in non-lymphoid tissue cells and tumor cells, in addition to tissue-recruiting immune cells, under inflammatory conditions triggered by several cytokines, especially IFN-γ, and exogenous stimuli delivered by pathogen-associated molecular patterns. Receptor-mediated signaling molecules that affect the cell cycle, proliferation, apoptosis, and survival (including NF-κB, MAPK, PI3K, mTOR, and JAK/STAT) are involved in PD-L1 induction. PD-L1 expression in tumor cells is also triggered by the signals described above, but in some instances, intrinsic cell alteration associated with carcinogenesis contributes to PD-L1 induction. The tumor suppressor genes PTEN and Lkb1 and epithelial-mesenchymal transition-related molecules are also involved in the regulation of PD-L1 expression. Notably, squamous cell carcinoma of the head and neck (SCCHN) often exhibits both host immunosuppression and cytogenetic alternations of tumor cells. Precise understanding of how PD-L1 expression is controlled will allow the development of effective approaches to PD-1 blockade therapy for patients with SCCHN.
Collapse
Affiliation(s)
- Patcharee Ritprajak
- Department of Microbiology and Immunology and DRU of Oral Microbiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Miyuki Azuma
- Department of Molecular Immunology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan.
| |
Collapse
|
139
|
Deng R, Cassady K, Li X, Yao S, Zhang M, Racine J, Lin J, Chen L, Zeng D. B7H1/CD80 interaction augments PD-1-dependent T cell apoptosis and ameliorates graft-versus-host disease. THE JOURNAL OF IMMUNOLOGY 2014; 194:560-74. [PMID: 25488990 DOI: 10.4049/jimmunol.1402157] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Interactions of B7H1 (programmed death ligand 1 [PD-L1]) with its two ligands, PD-1 and CD80, on T cells play a pivotal role in controlling T cell activation, proliferation, anergy, and apoptosis. However, the interactions between the two pathways remain unknown. Using an alloimmune response model of graft-versus-host disease (GVHD), we report in this study that: 1) Comparison of proliferation and apoptosis of wild-type (WT) and PD-1(-/-)CD4(+) conventional T (Tcon) cells in WT and B7H1(-/-) recipients revealed that B7H1/CD80 interaction per se augments T cell proliferation, and this interaction augments T cell apoptosis mediated by B7H1/PD-1 interaction. This observation was recapitulated in an in vitro MLR assay. 2) Specific blockade of the B7H1/CD80 axis by anti-B7H1 mAb reduces WT-alloreactive Tcon cell proliferation, IL-2 production, expression of PD-1, and apoptosis, resulting in worsening GVHD. In contrast, specific blockade of B7H1/CD80 interaction reduces donor PD-1(-/-) Tcon cell proliferation without an impact on apoptosis, resulting in ameliorating GVHD. 3) B7H1 fused to an Ig Fc domain (B7H1-Ig), when produced in vivo by hydrodynamic injection of B7H1-Ig plasmid, ameliorates GVHD by augmenting proliferation and apoptosis of WT- alloreactive Tcon cells. Conversely, B7H1-Ig treatment has no impact on apoptosis but augments PD-1(-/-) T cell proliferation and worsens GVHD. These results indicate that B7H1/CD80 interaction augments Tcon cell proliferation, IL-2 production, and expression of PD-1, which leads to increased apoptosis mediated by the B7H1/PD-1 pathway. Additionally, by engaging both PD-1 and CD80, B7H1-Ig can be a powerful therapeutic reagent for downregulating the T cell immune response.
Collapse
Affiliation(s)
- Ruishu Deng
- Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010
| | - Kaniel Cassady
- Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010; Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA 91010
| | - Xiaofan Li
- Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Sheng Yao
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520; and
| | - Mingfeng Zhang
- Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010
| | - Jeremy Racine
- Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010; Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA 91010
| | - Jeffrey Lin
- Eugene and Ruth Roberts Summer Student Academy of City of Hope, Duarte, CA 91010
| | - Lieping Chen
- Department of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Defu Zeng
- Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010; Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA 91010;
| |
Collapse
|
140
|
Glick GD, Rossignol R, Lyssiotis CA, Wahl D, Lesch C, Sanchez B, Liu X, Hao LY, Taylor C, Hurd A, Ferrara JLM, Tkachev V, Byersdorfer CA, Boros L, Opipari AW. Anaplerotic metabolism of alloreactive T cells provides a metabolic approach to treat graft-versus-host disease. J Pharmacol Exp Ther 2014; 351:298-307. [PMID: 25125579 PMCID: PMC4201277 DOI: 10.1124/jpet.114.218099] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 08/13/2014] [Indexed: 01/19/2023] Open
Abstract
T-cell activation requires increased ATP and biosynthesis to support proliferation and effector function. Most models of T-cell activation are based on in vitro culture systems and posit that aerobic glycolysis is employed to meet increased energetic and biosynthetic demands. By contrast, T cells activated in vivo by alloantigens in graft-versus-host disease (GVHD) increase mitochondrial oxygen consumption, fatty acid uptake, and oxidation, with small increases of glucose uptake and aerobic glycolysis. Here we show that these differences are not a consequence of alloactivation, because T cells activated in vitro either in a mixed lymphocyte reaction to the same alloantigens used in vivo or with agonistic anti-CD3/anti-CD28 antibodies increased aerobic glycolysis. Using targeted metabolic (13)C tracer fate associations, we elucidated the metabolic pathway(s) employed by alloreactive T cells in vivo that support this phenotype. We find that glutamine (Gln)-dependent tricarboxylic acid cycle anaplerosis is increased in alloreactive T cells and that Gln carbon contributes to ribose biosynthesis. Pharmacological modulation of oxidative phosphorylation rapidly reduces anaplerosis in alloreactive T cells and improves GVHD. On the basis of these data, we propose a model of T-cell metabolism that is relevant to activated lymphocytes in vivo, with implications for the discovery of new drugs for immune disorders.
Collapse
Affiliation(s)
- Gary D Glick
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Rodrigue Rossignol
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Costas A Lyssiotis
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Daniel Wahl
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Charles Lesch
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Brian Sanchez
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Xikui Liu
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Ling-Yang Hao
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Clarke Taylor
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Alexander Hurd
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - James L M Ferrara
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Victor Tkachev
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Craig A Byersdorfer
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Laszlo Boros
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Anthony W Opipari
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| |
Collapse
|
141
|
α-1-Antitrypsin (AAT)-modified donor cells suppress GVHD but enhance the GVL effect: a role for mitochondrial bioenergetics. Blood 2014; 124:2881-91. [PMID: 25224412 DOI: 10.1182/blood-2014-04-570440] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Hematopoietic cell transplantation is curative in many patients. However, graft-versus-host disease (GVHD), triggered by alloreactive donor cells, has remained a major complication. Here, we show an inverse correlation between plasma α-1-antitrypsin (AAT) levels in human donors and the development of acute GVHD in the recipients (n = 111; P = .0006). In murine models, treatment of transplant donors with human AAT resulted in an increase in interleukin-10 messenger RNA and CD8(+)CD11c(+)CD205(+) major histocompatibility complex class II(+) dendritic cells (DCs), and the prevention or attenuation of acute GVHD in the recipients. Ablation of DCs (in AAT-treated CD11c-DTR donors) decreased CD4(+)CD25(+)FoxP3(+) regulatory T cells to one-third and abrogated the anti-GVHD effect. The graft-versus-leukemia (GVL) effect of donor cells (against A20 tumor cells) was maintained or even enhanced with AAT treatment of the donor, mediated by an expanded population of NK1.1(+), CD49B(+), CD122(+), CD335(+) NKG2D-expressing natural killer (NK) cells. Blockade of NKG2D significantly suppressed the GVL effect. Metabolic analysis showed a high glycolysis-high oxidative phosphorylation profile for NK1.1(+) cells, CD4(+)CD25(+)FoxP3(+) T cells, and CD11c(+) DCs but not for effector T cells, suggesting a cell type-specific effect of AAT. Thus, via altered metabolism, AAT exerts effective GVHD protection while enhancing GVL effects.
Collapse
|
142
|
Host-derived CD8⁺ dendritic cells protect against acute graft-versus-host disease after experimental allogeneic bone marrow transplantation. Biol Blood Marrow Transplant 2014; 20:1696-704. [PMID: 25132527 DOI: 10.1016/j.bbmt.2014.08.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 08/06/2014] [Indexed: 11/22/2022]
Abstract
Graft-versus-host disease (GVHD) is a frequent life-threatening complication after allogeneic hematopoietic stem cell transplantation (HSCT) and induced by donor-derived T cells that become activated by host antigen-presenting cells. To address the relevance of host dendritic cell (DC) populations in this disease, we used mouse strains deficient in CD11c(+) or CD8α(+) DC populations in a model of acute GVHD where bone marrow and T cells from BALB/c donors were transplanted into C57BL/6 hosts. Surprisingly, a strong increase in GVHD-related mortality was observed in the absence of CD11c(+) cells. Likewise, Batf3-deficient (Batf3(-/-)) mice that lack CD8α(+) DCs also displayed a strongly increased GVHD-related mortality. In the absence of CD8α(+) DCs, we detected an increased activation of the remaining DC populations after HSCT, leading to an enhanced priming of allogeneic T cells. Importantly, this was associated with reduced numbers of regulatory T cells and transforming growth factor-β levels, indicating an aggravated failure of peripheral tolerance mechanisms after HSCT in the absence of CD8α(+) DCs. In summary, our results indicate a critical role of CD8α(+) DCs as important inducers of regulatory T cell-mediated tolerance to control DC activation and T cell priming in the initiation phase of GVHD.
Collapse
|
143
|
Fujiwara H, Maeda Y, Kobayashi K, Nishimori H, Matsuoka KI, Fujii N, Kondo E, Tanaka T, Chen L, Azuma M, Yagita H, Tanimoto M. Programmed death-1 pathway in host tissues ameliorates Th17/Th1-mediated experimental chronic graft-versus-host disease. THE JOURNAL OF IMMUNOLOGY 2014; 193:2565-73. [PMID: 25080485 DOI: 10.4049/jimmunol.1400954] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Chronic graft-versus-host disease (GVHD) is a major cause of late death and morbidity after allogeneic hematopoietic cell transplantation, but its pathogenesis remains unclear. We investigated the role of the programmed death-1 (PD-1) pathway in chronic GVHD using a well-defined mouse model of B10.D2 (H-2(d)) donor to BALB/c (H-2(d)) recipients. PD-1 expression on allogeneic donor T cells was upregulated continuously in chronic GVHD development, whereas PD-L1 expression in host tissues was transiently upregulated and declined to basal levels in the late posttransplant period. Blockade of the PD-1 pathway by anti-PD-1, anti-PD-L1, or anti-PD-L2 mAbs exacerbated clinical and pathologic chronic GVHD. Chimeric mice revealed that PD-L1 expression in host tissues suppressed expansion of IL-17(+)IFN-γ(+) T cells, and that PD-L1 expression on hematopoietic cells plays a role in the development of regulatory T cells only during the early transplantation period but does not affect the severity of chronic GVHD. Administration of the synthetic retinoid Am80 overcame the IL-17(+)IFN-γ(+) T cell expansion caused by PD-L1 deficiency, resulting in reduced chronic GVHD damage in PD-L1(-/-) recipients. Stimulation of the PD-1 pathway also alleviated chronic GVHD. These results suggest that the PD-1 pathway contributes to the suppression of Th17/Th1-mediated chronic GVHD and may represent a new target for the prevention or treatment of chronic GVHD.
Collapse
Affiliation(s)
- Hideaki Fujiwara
- Department of Hematology and Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan 700-8558
| | - Yoshinobu Maeda
- Department of Hematology and Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan 700-8558;
| | - Koichiro Kobayashi
- Department of Hematology and Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan 700-8558
| | - Hisakazu Nishimori
- Department of Hematology and Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan 700-8558
| | - Ken-Ichi Matsuoka
- Department of Hematology and Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan 700-8558
| | - Nobuharu Fujii
- Department of Hematology and Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan 700-8558
| | - Eisei Kondo
- Department of Hematology and Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan 700-8558
| | - Takehiro Tanaka
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan 700-8558
| | - Lieping Chen
- Department of Immunobiology and Yale Comprehensive Cancer Center, Yale University, New Haven, CT 06519
| | - Miyuki Azuma
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan 113-8549; and
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan 113-8421
| | - Mitsune Tanimoto
- Department of Hematology and Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan 700-8558
| |
Collapse
|
144
|
Okiyama N, Katz SI. Programmed cell death 1 (PD-1) regulates the effector function of CD8 T cells via PD-L1 expressed on target keratinocytes. J Autoimmun 2014; 53:1-9. [PMID: 25047812 DOI: 10.1016/j.jaut.2014.06.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 06/16/2014] [Accepted: 06/29/2014] [Indexed: 01/22/2023]
Abstract
Programmed cell death 1 (PD-1) is an inhibitory molecule expressed by activated T cells. Its ligands (PD-L1 and -L2; PD-Ls) are expressed not only by a variety of leukocytes but also by stromal cells. To assess the role of PD-1 in CD8 T cell-mediated diseases, we used PD-1-knockout (KO) OVA-specific T cell-receptor transgenic (Tg) CD8 T cells (OT-I cells) in a murine model of mucocutaneous graft-versus-host disease (GVHD). We found that mice expressing OVA on epidermal keratinocytes (K14-mOVA mice) developed markedly enhanced GVHD-like disease after transfer of PD-1-KO OT-I cells as compared to those mice transferred with wild-type OT-I cells. In addition, K14-mOVA × OT-I double Tg (DTg) mice do not develop GVHD-like disease after adoptive transfer of OT-I cells, while transfer of PD-1-KO OT-I cells caused GVHD-like disease in a Fas/Fas-L independent manner. These results suggest that PD-1/PD-Ls-interactions have stronger inhibitory effects on pathogenic CD8 T cells than does Fas/Fas-L-interactions. Keratinocytes from K14-mOVA mice with GVHD-like skin lesions express PD-L1, while those from mice without the disease do not. These findings reflect the fact that primary keratinocytes express PD-L1 when stimulated by interferon-γ in vitro. When co-cultured with K14-mOVA keratinocytes for 2 days, PD-1-KO OT-I cells exhibited enhanced proliferation and activation compared to wild-type OT-I cells. In addition, knockdown of 50% PD-L1 expression on the keratinocytes with transfection of PD-L1-siRNA enhanced OT-I cell proliferation. In aggregate, our data strongly suggest that PD-L1, expressed on activated target keratinocytes presenting autoantigens, regulates autoaggressive CD8 T cells, and inhibits the development of mucocutaneous autoimmune diseases.
Collapse
Affiliation(s)
- Naoko Okiyama
- Dermatology Branch, National Cancer Institute, National Institutes of Health, Bldg 10, 12N250, 10 Center Dr, Bethesda, MD 20892, USA.
| | - Stephen I Katz
- Dermatology Branch, National Cancer Institute, National Institutes of Health, Bldg 10, 12N250, 10 Center Dr, Bethesda, MD 20892, USA.
| |
Collapse
|
145
|
Macintyre AN, Gerriets VA, Nichols AG, Michalek RD, Rudolph MC, Deoliveira D, Anderson SM, Abel ED, Chen BJ, Hale LP, Rathmell JC. The glucose transporter Glut1 is selectively essential for CD4 T cell activation and effector function. Cell Metab 2014; 20:61-72. [PMID: 24930970 PMCID: PMC4079750 DOI: 10.1016/j.cmet.2014.05.004] [Citation(s) in RCA: 878] [Impact Index Per Article: 79.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 12/11/2013] [Accepted: 04/24/2014] [Indexed: 12/13/2022]
Abstract
CD4 T cell activation leads to proliferation and differentiation into effector (Teff) or regulatory (Treg) cells that mediate or control immunity. While each subset prefers distinct glycolytic or oxidative metabolic programs in vitro, requirements and mechanisms that control T cell glucose uptake and metabolism in vivo are uncertain. Despite expression of multiple glucose transporters, Glut1 deficiency selectively impaired metabolism and function of thymocytes and Teff. Resting T cells were normal until activated, when Glut1 deficiency prevented increased glucose uptake and glycolysis, growth, proliferation, and decreased Teff survival and differentiation. Importantly, Glut1 deficiency decreased Teff expansion and the ability to induce inflammatory disease in vivo. Treg cells, in contrast, were enriched in vivo and appeared functionally unaffected and able to suppress Teff, irrespective of Glut1 expression. These data show a selective in vivo requirement for Glut1 in metabolic reprogramming of CD4 T cell activation and Teff expansion and survival.
Collapse
Affiliation(s)
- Andrew N Macintyre
- Department of Pharmacology and Cancer Biology, Immunology, Sarah W. Stedman Nutrition and Metabolism Center, Duke University, Durham, NC 27710, USA
| | - Valerie A Gerriets
- Department of Pharmacology and Cancer Biology, Immunology, Sarah W. Stedman Nutrition and Metabolism Center, Duke University, Durham, NC 27710, USA
| | - Amanda G Nichols
- Department of Pharmacology and Cancer Biology, Immunology, Sarah W. Stedman Nutrition and Metabolism Center, Duke University, Durham, NC 27710, USA
| | - Ryan D Michalek
- Department of Pharmacology and Cancer Biology, Immunology, Sarah W. Stedman Nutrition and Metabolism Center, Duke University, Durham, NC 27710, USA
| | - Michael C Rudolph
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Divino Deoliveira
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke University, Durham, NC 27710, USA
| | - Steven M Anderson
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - E Dale Abel
- Fraternal Order of Eagles Diabetes Research Center, Division of Endocrinology and Metabolism, Department of Medicine, Carver College of Medicine University of Iowa, Iowa City, IA 52242, USA
| | - Benny J Chen
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke University, Durham, NC 27710, USA
| | - Laura P Hale
- Department of Pathology, Duke University, Durham, NC 27710, USA
| | - Jeffrey C Rathmell
- Department of Pharmacology and Cancer Biology, Immunology, Sarah W. Stedman Nutrition and Metabolism Center, Duke University, Durham, NC 27710, USA
| |
Collapse
|
146
|
|