51
|
Thummalapalli R, Sena LA, Probasco JC, Gladstone DE. Checkpoint inhibitor-induced autoimmune encephalitis reversed by rituximab after allogeneic bone marrow transplant in a patient with Hodgkin lymphoma. Leuk Lymphoma 2019; 61:228-230. [DOI: 10.1080/10428194.2019.1658104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
| | - Laura A. Sena
- Department of Medical Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - John C. Probasco
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Douglas E. Gladstone
- Department of Medical Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| |
Collapse
|
52
|
Immune checkpoint inhibitors as a bridge to allogeneic transplantation with posttransplant cyclophosphamide. Blood Adv 2019; 2:2226-2229. [PMID: 30190282 DOI: 10.1182/bloodadvances.2018019208] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 08/06/2018] [Indexed: 01/15/2023] Open
Abstract
Published reports suggest that immune checkpoint inhibitors (ICIs) before allogeneic blood or marrow transplantation (alloBMT) may increase the incidence of graft-versus-host disease (GvHD), immune-related adverse events, and nonrelapse mortality (NRM); this led to the US Food and Drug Administration issuing a "Warning and Precaution" regarding the potential for life-threatening immune-mediated complications associated with alloBMT after nivolumab and pembrolizumab. We retrospectively reviewed the outcomes of 14 consecutive patients who received ICIs as their final salvage therapy before T-cell-replete alloBMT using reduced-intensity conditioning. All patients received posttransplant cyclophosphamide (PTCy), which significantly limits severe GvHD, even in the mismatched-donor setting. There was no grade 3-4 acute GvHD (aGvHD), and all 6 cases of grade 2 aGvHD readily resolved with immunosuppression. No patient experienced veno-occlusive disease of the liver, other immune-related adverse events, chronic GvHD, or NRM. There have been 2 relapses (15-month median follow-up), with 12 of 14 patients remaining alive, well, and progression-free. The only death was a result of disease relapse. Although more experience is needed, our data suggest that concerns over immunologic complications associated with ICIs should not preclude allogeneic bone marrow transplantation with PTCy as GvHD prophylaxis.
Collapse
|
53
|
Holtan SG, Shabaneh A, Betts BC, Rashidi A, MacMillan ML, Ustun C, Amin K, Vaughn BP, Howard J, Khoruts A, Arora M, DeFor TE, Johnson D, Blazar BR, Weisdorf DJ, Wang J. Stress responses, M2 macrophages, and a distinct microbial signature in fatal intestinal acute graft-versus-host disease. JCI Insight 2019; 5:129762. [PMID: 31393854 DOI: 10.1172/jci.insight.129762] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Steroid-refractory intestinal acute graft-versus-host disease (aGVHD) is a frequently fatal condition with little known about mechanisms driving failed steroid responses in gut mucosa. To uncover novel molecular insights in steroid-refractory aGVHD, we compared gene expression profiles of rectosigmoid biopsies from patients at diagnosis of clinical stage 3-4 lower intestinal aGVHD (N=22), to repeat biopsies when the patients became steroid refractory (N=22), and normal controls (N=10). We also performed single gene analyses of factors associated with tolerance (programmed death ligand-1 [PDL1], indoleamine 2,3 dioxygenase [IDO1], and T cell immunoreceptor with Ig and ITIM domains [TIGIT]) and found that significantly higher expression levels of these aGVHD inhibitory genes (PDL1, IDO1, TIGIT) at aGVHD onset became decreased in the steroid-refractory state. We examined genes triggered by microbial ligands to stimulate gut repair, amphiregulin (AREG) and the aryl hydrocarbon receptor (AhR), and found that both AREG and AhR gene expression levels were increased at aGVHD onset and remained elevated in steroid-refractory aGVHD. We also identified higher expression levels of metallothioneines, metal-binding enzymes induced in stress responses, and M2 macrophage genes in steroid-refractory aGVHD. We observed no differences in T-cell subsets between onset and steroid-refractory aGVHD. Patients with a rapidly fatal course showed greater DNA damage and a distinct microbial signature at aGVHD onset, whereas patients with more prolonged survival exhibited a gene expression profile consistent with activation of Smoothened. Our results extend the paradigm beyond T cell-centric therapies for steroid-refractory GI aGVHD and highlight new mechanisms for therapeutic exploration.
Collapse
Affiliation(s)
| | | | - Brian C Betts
- Blood and Marrow Transplant Program, Department of Medicine
| | - Armin Rashidi
- Blood and Marrow Transplant Program, Department of Medicine
| | - Margaret L MacMillan
- Blood and Marrow Transplant Program, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Celalletin Ustun
- Rush University Blood and Marrow Transplant Program, Chicago, Illinois, USA
| | | | | | - Justin Howard
- Division of Gastroenterology, Department of Medicine
| | | | - Mukta Arora
- Blood and Marrow Transplant Program, Department of Medicine
| | | | | | - Bruce R Blazar
- Blood and Marrow Transplant Program, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Jinhua Wang
- Cancer Bioinformatics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
54
|
Brodská B, Otevřelová P, Šálek C, Fuchs O, Gašová Z, Kuželová K. High PD-L1 Expression Predicts for Worse Outcome of Leukemia Patients with Concomitant NPM1 and FLT3 Mutations. Int J Mol Sci 2019; 20:ijms20112823. [PMID: 31185600 PMCID: PMC6600137 DOI: 10.3390/ijms20112823] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/31/2019] [Accepted: 06/06/2019] [Indexed: 12/23/2022] Open
Abstract
Compared to solid tumors, the role of PD-L1 in hematological malignancies is less explored, and the knowledge in this area is mostly limited to lymphomas. However, several studies indicated that PD-L1 is also overexpressed in myeloid malignancies. Successful treatment of the acute myeloid leukemia (AML) is likely associated with elimination of the residual disease by the immune system, and possible involvement of PD-L1 in this process remains to be elucidated. We analyzed PD-L1 expression on AML primary cells by flow cytometry and, in parallel, transcript levels were determined for the transcription variants v1 and v2. The ratio of v1/v2 cDNA correlated with the surface protein amount, and high v1/v2 levels were associated with worse overall survival (p = 0.0045). The prognostic impact of PD-L1 was limited to AML with mutated nucleophosmin and concomitant internal tandem duplications in the FLT3 gene (p less than 0.0001 for this particular AML subgroup).
Collapse
Affiliation(s)
- Barbora Brodská
- Institute of Hematology and Blood Transfusion, Prague 128 20, Czech Republic.
| | - Petra Otevřelová
- Institute of Hematology and Blood Transfusion, Prague 128 20, Czech Republic.
| | - Cyril Šálek
- Institute of Hematology and Blood Transfusion, Prague 128 20, Czech Republic.
| | - Ota Fuchs
- Institute of Hematology and Blood Transfusion, Prague 128 20, Czech Republic.
| | - Zdenka Gašová
- Institute of Hematology and Blood Transfusion, Prague 128 20, Czech Republic.
| | - Kateřina Kuželová
- Institute of Hematology and Blood Transfusion, Prague 128 20, Czech Republic.
| |
Collapse
|
55
|
Immune Checkpoint Inhibition in Classical Hodgkin Lymphoma: From Early Achievements towards New Perspectives. JOURNAL OF ONCOLOGY 2019; 2019:9513701. [PMID: 31205470 PMCID: PMC6530161 DOI: 10.1155/2019/9513701] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 03/18/2019] [Accepted: 04/17/2019] [Indexed: 02/07/2023]
Abstract
Immune checkpoint inhibition (ICI) became one of the major breakthroughs in cancer treatment over the past decade and entered into therapy within standard oncohematology practice. ICI has demonstrated impressive response rates as salvage therapy in relapsed/refractory (R/R) classical Hodgkin lymphoma (cHL) and is now being tested as an adjunction to chemotherapy in the frontline settings. CHL exquisite sensitivity to PD-1/PD-L1 axis inhibition relies on a particular biological background. By contrast, non-Hodgkin lymphomas (NHL) have demonstrated heterogeneous response rates using ICI. These observations highlight discrepancies between various types of lymphomas in terms of genetic alterations, immune microenvironment interactions, and disease phenotype. This review aims to focus on cHL immune escape mechanisms, focusing on cHL biological sensitivity to PD-1 blockade. We will summarize the available data issued from clinical trials on ICI in cHL and its safety profile. Going beyond the current use of monoclonal antibodies (mAb) targeting immune checkpoints in clinical practice, we will offer an overview of new combinatory therapeutic perspectives where cHL immunotherapy may be considered.
Collapse
|
56
|
Tang L, Ma S, Gong H, Wang J, Xu Y, Wu D, Sun A. PD-L1 Ameliorates Murine Acute Graft-Versus-Host Disease by Suppressing Effector But Not Regulatory T Cells Function. Arch Immunol Ther Exp (Warsz) 2019; 67:179-187. [PMID: 30927016 DOI: 10.1007/s00005-019-00539-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 03/18/2019] [Indexed: 01/09/2023]
Abstract
There is increasing evidence that interaction between programmed death 1 (PD-1) and its ligands PD-1 (PD-L1) plays a critical role in the pathology of acute graft-versus-host disease (aGVHD). However, the role of PD-L1 in the development of aGVHD has been controversial in recent mouse studies. In this study, we carried out studies in a murine aGVHD model to clarify the role of PD-L1 in aGVHD pathogenesis. We found that systemic overexpression of PD-L1 by hydrodynamic gene transfer (HGT) method in vivo ameliorates aGVHD-induced lethality in mice. Systemic overexpression of PD-L1 inhibits the donor T cells activation, effector memory status, as well as Th1 and Th17 cells responses in vivo. In addition, PD-L1 Ig treatment significantly suppressed T cells' proliferation, promoted T cells' apoptosis, and reduced pro-inflammatory cytokines expression by effector T cells in vitro in the stimulation of anti-CD3/CD28 and allogeneic dendritic cells. However, we found that PD-L1 overexpression did not affect Treg cells' differentiation in vivo and in vitro, depletion of Treg cells in PD-L1 HGT recipients did not aggravate aGVHD mortality. Therefore, our results demonstrated that systemic treatment with PD-L1 protein ameliorates aGVHD by suppressing effector but not regulatory T cell function. Our findings suggest that systemic treatment with PD-L1 may be a potential strategy to prevent or ameliorate aGVHD.
Collapse
Affiliation(s)
- Lin Tang
- Department of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.,Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, 215123, China.,Department of Hematology, Changzhou Traditional Chinese Medicine Hospital, Changzhou, 213003, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006, China
| | - Shoubao Ma
- Department of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China. .,Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, 215123, China. .,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006, China.
| | - Huanle Gong
- Department of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.,Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, 215123, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006, China
| | - Jun Wang
- Department of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.,Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, 215123, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006, China
| | - Yang Xu
- Department of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.,Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, 215123, China.,Department of Hematology, Changzhou Traditional Chinese Medicine Hospital, Changzhou, 213003, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006, China
| | - Depei Wu
- Department of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China. .,Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, 215123, China. .,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006, China.
| | - Aining Sun
- Department of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China. .,Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, 215123, China. .,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006, China.
| |
Collapse
|
57
|
Thangavelu G, Blazar BR. Achievement of Tolerance Induction to Prevent Acute Graft-vs.-Host Disease. Front Immunol 2019; 10:309. [PMID: 30906290 PMCID: PMC6419712 DOI: 10.3389/fimmu.2019.00309] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 02/06/2019] [Indexed: 01/04/2023] Open
Abstract
Acute graft-vs.-host disease (GVHD) limits the efficacy of allogeneic hematopoietic stem cell transplantation (allo-HSCT), a main therapy to treat various hematological disorders. Despite rapid progress in understanding GVHD pathogenesis, broad immunosuppressive agents are most often used to prevent and remain the first line of therapy to treat GVHD. Strategies enhancing immune tolerance in allo-HSCT would permit reductions in immunosuppressant use and their associated undesirable side effects. In this review, we discuss the mechanisms responsible for GVHD and advancement in strategies to achieve immune balance and tolerance thereby avoiding GVHD and its complications.
Collapse
Affiliation(s)
- Govindarajan Thangavelu
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
58
|
The co-inhibitory molecule PD-L1 contributes to regulatory T cell-mediated protection in murine crescentic glomerulonephritis. Sci Rep 2019; 9:2038. [PMID: 30765734 PMCID: PMC6375967 DOI: 10.1038/s41598-018-38432-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 12/19/2018] [Indexed: 12/15/2022] Open
Abstract
Immune-mediated glomerular diseases like crescentic glomerulonephritis (cGN) are driven by inappropriately regulated cellular and humoral immune responses subsequently leading to renal tissue injury. Recent studies demonstrated the crucial role for regulatory T cells (Tregs) in suppressing pathogenic T-cell responses during nephrotoxic nephritis (NTN), a murine model of cGN. However, mechanisms of immune regulation in cGN are less clear. Here, we aim at investigating the role of the co-inhibitory PD-1/PD-L1 pathway in Treg-mediated suppression of renal inflammation. We demonstrated that Foxp3+ Tregs expressing PD-L1 infiltrate the kidney during NTN. Inhibition of PD-L1 signalling by using PD-L1−/− mice or by blockage of PD-L1 in wildtype mice resulted in an increased Treg frequency in the inflamed kidney. However, mice lacking PD-L1 developed more severe NTN associated with an elevated pathogenic renal Th1 immune response, which was reversed by blockage of IFNγ in these mice. Interestingly, lack of PD-L1 altered the gene expression profile of Tregs in homeostasis and kidney inflammation. Functionally, Tregs from nephritic PD-L1−/− mice had impaired suppressive capacity in vitro and failed to protect from NTN in vivo. Thus, PD-L1 displays a protective role in NTN, which is related to Treg-mediated suppression of the Th1 immune response.
Collapse
|
59
|
Cassady K, Martin PJ, Zeng D. Regulation of GVHD and GVL Activity via PD-L1 Interaction With PD-1 and CD80. Front Immunol 2018; 9:3061. [PMID: 30622541 PMCID: PMC6308317 DOI: 10.3389/fimmu.2018.03061] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/10/2018] [Indexed: 12/18/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (HCT) is a curative therapy for hematological malignancies (i.e. leukemia and lymphoma), because graft-versus-leukemia (GVL) activity mediated by alloreactive T cells can eliminate residual malignant cells and prevent relapse. However, the same alloreactive T cells also mediate a severe side effect, graft-versus-host disease (GVHD), and prevention of GVHD while preserving GVL activity remains an elusive goal. The immune checkpoint molecule PD-L1 and its interaction with PD-1 receptor in regulating cancer immunity is under intensive and wide-spread study, but knowledge about this interaction in regulating GVHD and GVL activity is very limited. In this review, we summarize the literature exploring how PD-L1 interaction with its receptors PD-1 and CD80 regulate GVHD and GVL activities, how PD-L1 signaling regulates T cell metabolic profiles, and how a differential role of PD-L1 interaction with PD-1, CD80 or both may provide a novel avenue to prevent GVHD while preserving strong GVL effects.
Collapse
Affiliation(s)
- Kaniel Cassady
- Irell and Manella Graduate School of Biological Sciences of City of Hope, Duarte, CA, United States.,Department of Hematology/Hematopoietic Cell Transplantation, Beckman Research Institute at City of Hope National Medical Center, Duarte, CA, United States.,Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Paul J Martin
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, United States.,Department of Medicine, University of Washington, Seattle, WA, United States
| | - Defu Zeng
- Irell and Manella Graduate School of Biological Sciences of City of Hope, Duarte, CA, United States.,Department of Hematology/Hematopoietic Cell Transplantation, Beckman Research Institute at City of Hope National Medical Center, Duarte, CA, United States.,Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| |
Collapse
|
60
|
Du W, Cao X. Cytotoxic Pathways in Allogeneic Hematopoietic Cell Transplantation. Front Immunol 2018; 9:2979. [PMID: 30631325 PMCID: PMC6315278 DOI: 10.3389/fimmu.2018.02979] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/04/2018] [Indexed: 12/11/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is a potentially curative treatment for hematologic malignancies, and other hematologic and immunologic diseases. Donor-derived immune cells identify and attack cancer cells in the patient producing a unique graft-vs.-tumor (GVT) effect. This beneficial response renders allo-HCT one of the most effective forms of tumor immunotherapy. However, alloreactive donor T cells can damage normal host cells thereby causing graft-vs.-host disease (GVHD), which results in substantial morbidity and mortality. To date, GVHD remains as the major obstacle for more successful application of allo-HCT. Of special significance in this context are a number of cytotoxic pathways that are involved in GVHD and GVT response as well as donor cell engraftment. In this review, we summarize progress in the investigation of these cytotoxic pathways, including Fas/Fas ligand (FasL), perforin/granzyme, and cytokine pathways. Many studies have delineated their distinct operating mechanisms and how they are involved in the complex cellular interactions amongst donor, host, tumor, and infectious pathogens. Driven by progressing elucidation of their contributions in immune reconstitution and regulation, various interventional strategies targeting these pathways have entered translational stages with aims to improve the effectiveness of allo-HCT.
Collapse
Affiliation(s)
- Wei Du
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Xuefang Cao
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States.,Department of Microbiology and Immunology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
61
|
Abstract
Background Checkpoint inhibitors have transformed the treatment of cancer in adults. This class of drugs has demonstrated encouraging results in various malignancies such as metastatic melanoma, bladder cancer, renal cancer, and non-small cell lung carcinoma. However, researchers have only begun investigating the effectiveness and tolerability of checkpoint inhibitors in pediatric patients. Methods We conducted a review of PubMed indexed literature and clinicaltrials.gov using combinations of the keywords checkpoint, inhibitor, pediatric, CTLA-4 (cytotoxic T lymphocyte antigen-4), PD-1 (programmed cell death-1), and PD-L1 (programmed cell death receptor-1 ligand) to find every recently completed and ongoing trial evaluating checkpoint inhibitors in patients younger than 21 years old. Pertinent articles and clinical trials discussing the role of immune checkpoint inhibitors in the pediatric population were selected for final analysis and manuscript citation. Results This review presents an overview of the cellular mechanisms involved in checkpoint inhibition and of studies evaluating checkpoint inhibitors in humans. The review also details results and side effects from studies conducted with pediatric patients, current pediatric clinical trials, and future implications. Conclusion Immune checkpoint inhibitors have the potential to further therapeutic advances in pediatric oncology; however, we need more clinical trials and combination drug strategies targeted toward pediatric cancers.
Collapse
|
62
|
Choi J, Cooper ML, Staser K, Ashami K, Vij KR, Wang B, Marsala L, Niswonger J, Ritchey J, Alahmari B, Achilefu S, Tsunoda I, Schroeder MA, DiPersio JF. Baricitinib-induced blockade of interferon gamma receptor and interleukin-6 receptor for the prevention and treatment of graft-versus-host disease. Leukemia 2018; 32:2483-2494. [PMID: 29691471 PMCID: PMC6168427 DOI: 10.1038/s41375-018-0123-z] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/26/2018] [Accepted: 03/15/2018] [Indexed: 02/08/2023]
Abstract
The therapeutic benefits of allogeneic hematopoietic stem cell transplantation (allo-HSCT) are derived from the graft-versus-leukemia (GvL) effects of the procedure. There is a strong association between the GvL effects and graft-versus-host disease (GvHD), a major life-threatening complication of allo-HSCT. The limiting of GvHD while maintaining the GvL effect remains the goal of allo-HSCT. Therefore, identifying optimal therapeutic targets to selectively suppress GvHD while maintaining the GvL effects represents a significant unmet medical need. We demonstrate that the dual inhibition of interferon gamma receptor (IFNγR) and interleukin-6 receptor (IL6R) results in near-complete elimination of GvHD in a fully major histocompatibility complex-mismatched allo-HSCT model. Furthermore, baricitinib (an inhibitor of Janus kinases 1 and 2 (JAK1/JAK2) downstream of IFNγR/IL6R) completely prevented GvHD; expanded regulatory T cells by preserving JAK3-STAT5 signaling; downregulated CXCR3 and helper T cells 1 and 2 while preserving allogeneic antigen-presenting cell-stimulated T-cell proliferation; and suppressed the expression of major histocompatibility complex II (I-Ad), CD80/86, and PD-L1 on host antigen-presenting cells. Baricitinib also reversed established GvHD with 100% survival, thus demonstrating both preventive and therapeutic roles for this compound. Remarkably, baricitinib enhanced the GvL effects, possibly by downregulating tumor PD-L1 expression.
Collapse
Affiliation(s)
- Jaebok Choi
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Matthew L Cooper
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Karl Staser
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Medicine, Division of Dermatology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Kidist Ashami
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Kiran R Vij
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Bing Wang
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Lynne Marsala
- Department of Radiology, Molecular Imaging Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jessica Niswonger
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Julie Ritchey
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Bader Alahmari
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Samuel Achilefu
- Department of Radiology, Molecular Imaging Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ikuo Tsunoda
- Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan
| | - Mark A Schroeder
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - John F DiPersio
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
63
|
Ijaz A, Khan AY, Malik SU, Faridi W, Fraz MA, Usman M, Tariq MJ, Durer S, Durer C, Russ A, Parr NNC, Baig Z, Sagar F, Ali Z, McBride A, Anwer F. Significant Risk of Graft-versus-Host Disease with Exposure to Checkpoint Inhibitors before and after Allogeneic Transplantation. Biol Blood Marrow Transplant 2018; 25:94-99. [PMID: 30195074 DOI: 10.1016/j.bbmt.2018.08.028] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/28/2018] [Indexed: 12/17/2022]
Abstract
Investigators are using checkpoint inhibitors (CPIs) to treat aggressive hematologic malignancies in patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) and in some patients with relapsed disease after allo-HSCT. CTLA-4 inhibitors and PD-1 inhibitors are 2 main types of CPIs, which work through activation of the immune system. On one hand, CPIs can achieve graft-versus-tumor effect, and on the other hand, there is a risk of graft-versus-host disease (GVHD). After a comprehensive literature review, we included data (n = 283) from 24 studies (11 original manuscripts and 13 case reports or case series) and evaluated the results to assess the safety and efficacy of CPI use in conjunction with allo-HSCT. Among the 283 patients, 107 received CPI before allo-HSCT, and 176 received CPI after allo-HSCT. The most common indication for CPI use was for Hodgkin lymphoma. The CPIs used in various studies included ipilimumab, nivolumab, and pembrolizumab. Among the patients exposed to CPI before allo-HSCT, 56% developed acute GVHD and 29% developed chronic GVHD. Investigators reported 20 deaths, 60% of which were GVHD-related. The overall mortality risk with GVHD is 11%. In this group, investigators noted an objective response rate (ORR) in 68% of patients, with complete remission (CR) in 47%, partial remission (PR) in 21%, and stable disease in 11%. Among the patients who received a CPI after allo-HSCT for disease relapse, 14% developed acute GVHD and 9% developed chronic GVHD. Investigators reported 40 deaths, 28% of which were GVHD-related. The mortality risk with GVHD is approximately 7%. Investigators reported ORR in 54% of patients, with CR in 33%, PR in 21%, and disease stabilization in 5%. After careful evaluation of collective data, we found that CPI use both before and after allo-HSCT can be highly effective, but exposure can lead to a significantly increased risk of GVHD-related morbidity and mortality in this patient population. Despite limited availability of data, there is need for extreme caution while making decisions regarding the use of CPIs. Detailed discussions and prospective well-designed clinical trials are needed to explore this issue further.
Collapse
Affiliation(s)
- Awais Ijaz
- Department of Medicine, Hematology/Oncology, Blood and Marrow Transplantation, The University of Arizona, Tucson, Arizona
| | - Ali Younas Khan
- Department of Medicine, Hematology/Oncology, Blood and Marrow Transplantation, The University of Arizona, Tucson, Arizona
| | - Saad Ullah Malik
- Department of Medicine, Hematology/Oncology, Blood and Marrow Transplantation, The University of Arizona, Tucson, Arizona
| | - Warda Faridi
- Department of Medicine, Hematology/Oncology, Blood and Marrow Transplantation, The University of Arizona, Tucson, Arizona
| | - Muhammad Asad Fraz
- Department of Medicine, Hematology/Oncology, Blood and Marrow Transplantation, The University of Arizona, Tucson, Arizona
| | - Muhammad Usman
- Department of Medicine, Hematology/Oncology, Blood and Marrow Transplantation, The University of Arizona, Tucson, Arizona
| | - Muhammad Junaid Tariq
- Department of Medicine, Hematology/Oncology, Blood and Marrow Transplantation, The University of Arizona, Tucson, Arizona
| | - Seren Durer
- Department of Medicine, Hematology/Oncology, Blood and Marrow Transplantation, The University of Arizona, Tucson, Arizona
| | - Ceren Durer
- Department of Medicine, Hematology/Oncology, Blood and Marrow Transplantation, The University of Arizona, Tucson, Arizona
| | - Atlantis Russ
- Internal Medicine Residency Program, College of Medicine, The University of Arizona, Tucson, Arizona
| | | | - Zeeshan Baig
- Department of Internal Medicine, Hospital Medicine, Summit Medical Group, Summit, New Jersey
| | - Fnu Sagar
- Department of Medicine, Hematology/Oncology, Blood and Marrow Transplantation, The University of Arizona, Tucson, Arizona
| | - Zeeshan Ali
- Department of Internal Medicine, The University of Arizona, Tucson, Arizona
| | - Ali McBride
- Department of Pharmacy, The University of Arizona, Tucson, Arizona
| | - Faiz Anwer
- Department of Medicine, Hematology/Oncology, Blood and Marrow Transplantation, The University of Arizona, Tucson, Arizona; Department of Hematology, Medical Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio.
| |
Collapse
|
64
|
Xiao H, Li L, Pang Y, Wu Y, Jiang Z, Liu Z, Wu J, Xiao Y, Huang F, Liu Q, Zhang H, Luo Y, Huang H. Sequential treatment combining cladribine-based re-induction, myeloablative allogeneic HSCT, and prophylactic donor lymphocyte infusion: a promising treatment for refractory acute myeloid leukemia. Ann Hematol 2018; 97:2479-2490. [PMID: 30078144 DOI: 10.1007/s00277-018-3453-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 07/19/2018] [Indexed: 11/26/2022]
Abstract
We describe the first multicenter prospective study to assess the efficacy, safety, and immune reconstitution of a novel sequential transplant approach in 24 patients with primary induction failure/relapsed acute myeloid leukemia (AML). The sequential regimen consisted of cladribine 5 mg/m2/day and cytarabine 2 g/m2/day for 5 days and mitoxantrone 7 mg/m2/day for 3 days, followed by myeloablative allogeneic hematopoietic stem cell transplantation (allo-HSCT) using intravenous busulfan (3.2 mg/kg/day) for 4 days and cyclophosphamide (60 mg/kg/day) for 2 days. Patients in CR without acute graft-versus-host disease at day + 90 received prophylactic donor lymphocyte infusion (pDLI). At the time of transplantation, a marrow blast infiltration > 20% or any level of circulating blasts was found in 62.5% of patients. The cumulative incidence of relapse at 2 years was 29.8%. Overall survival (OS) was 74.5% at 1 year and 56.5% at 2 years. Leukemia-free survival (LFS) at 1 and 2 years was 62.5 and 50.5%, respectively. Multivariate analysis demonstrated that haploidentical related donor, pDLI, and experiencing chronic graft-versus-host disease (cGVHD) were protective from relapse. Total T cells and T cell subsets in peripheral blood recovered at 3 months post-HSCT. The expressions of immune checkpoints (cytotoxic T lymphocyte antigen 4 and programmed death 1) were extremely low in T cells over the first 1 year post-transplantation.
Collapse
Affiliation(s)
- Haowen Xiao
- Department of Hematology, Guangzhou General Hospital of Guangzhou Military Command, No. 111 Liuhua Rd., Guangzhou, 510010, Guangdong Province, People's Republic of China.
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China.
| | - Li Li
- Center of Cell-Biological Therapy and Research, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, Guangdong Province, People's Republic of China
| | - Yan Pang
- Department of Hematology, Guangzhou General Hospital of Guangzhou Military Command, No. 111 Liuhua Rd., Guangzhou, 510010, Guangdong Province, People's Republic of China
| | - Yuanbin Wu
- Department of Hematology, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, People's Republic of China
| | - Zujun Jiang
- Department of Hematology, Guangzhou General Hospital of Guangzhou Military Command, No. 111 Liuhua Rd., Guangzhou, 510010, Guangdong Province, People's Republic of China
| | - Zenghui Liu
- Department of Hematology, Guangzhou General Hospital of Guangzhou Military Command, No. 111 Liuhua Rd., Guangzhou, 510010, Guangdong Province, People's Republic of China
| | - Jiulong Wu
- Center of Cell-Biological Therapy and Research, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, Guangdong Province, People's Republic of China
| | - Yang Xiao
- Department of Hematology, Guangzhou General Hospital of Guangzhou Military Command, No. 111 Liuhua Rd., Guangzhou, 510010, Guangdong Province, People's Republic of China
| | - Fen Huang
- Department of Hematology, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Hang Zhang
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Yi Luo
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| |
Collapse
|
65
|
Recommendations for managing PD-1 blockade in the context of allogeneic HCT in Hodgkin lymphoma: taming a necessary evil. Blood 2018; 132:9-16. [DOI: 10.1182/blood-2018-02-811174] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 04/27/2018] [Indexed: 12/15/2022] Open
Abstract
Abstract
PD-1 blockade is an effective therapy in relapsed/refractory (R/R) classical Hodgkin Lymphoma (cHL) who have relapsed after or are ineligible for autologous hematopoietic cell transplantation (HCT). Although single-agent anti-PD-1 monoclonal antibodies (mAb’s) are associated with high response rates and durable remissions, available results to date suggest that a large majority of patients will eventually progress on therapy. Many of these patients are potential candidates for allogeneic HCT (allo-HCT) after receiving anti-PD-1 mAb’s, and allo-HCT remains for now the only treatment with demonstrated curative potential in this setting. However, initial reports suggested that allo-HCT in this setting may be associated with increased risk of early transplant-related toxicity, likely driven by lingering effects of PD-1 blockade. Furthermore, many patients with R/R cHL who undergo allo-HCT will relapse after transplantation, most often with limited treatment options. Here again, PD-1 blockade appears to yield high response rates, but with an increased risk of attendant immune toxicity. Many questions remain regarding the use of PD-1 blockade before or after allo-HCT, especially in relation to the feasibility, outcome, optimal timing, and method of allo-HCT after PD-1 blockade. Despite the scarcity of prospective data, these questions are unavoidable and must be tackled by clinicians in the routine care of patients with advanced cHL. We provide consensus recommendations of a working group based on available data and experience, in an effort to help guide treatment decisions until more definitive data are obtained.
Collapse
|
66
|
Phase 1 clinical trial evaluating abatacept in patients with steroid-refractory chronic graft-versus-host disease. Blood 2018; 131:2836-2845. [DOI: 10.1182/blood-2017-05-780239] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 03/07/2018] [Indexed: 12/11/2022] Open
Abstract
Key Points
Costimulatory blockade using abatacept represents a novel therapeutic approach for the treatment of cGVHD. Abatacept resulted in a clinical response in 44% of patients with both decreased prednisone use and T-cell PD-1 expression in responders.
Collapse
|
67
|
Bettonville M, d'Aria S, Weatherly K, Porporato PE, Zhang J, Bousbata S, Sonveaux P, Braun MY. Long-term antigen exposure irreversibly modifies metabolic requirements for T cell function. eLife 2018; 7:30938. [PMID: 29911570 PMCID: PMC6025959 DOI: 10.7554/elife.30938] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 06/14/2018] [Indexed: 12/11/2022] Open
Abstract
Energy metabolism is essential for T cell function. However, how persistent antigenic stimulation affects T cell metabolism is unknown. Here, we report that long-term in vivo antigenic exposure induced a specific deficit in numerous metabolic enzymes. Accordingly, T cells exhibited low basal glycolytic flux and limited respiratory capacity. Strikingly, blockade of inhibitory receptor PD-1 stimulated the production of IFNγ in chronic T cells, but failed to shift their metabolism towards aerobic glycolysis, as observed in effector T cells. Instead, chronic T cells appeared to rely on oxidative phosphorylation (OXPHOS) and fatty acid oxidation (FAO) to produce ATP for IFNγ synthesis. Check-point blockade, however, increased mitochondrial production of superoxide and reduced viability and effector function. Thus, in the absence of a glycolytic switch, PD-1-mediated inhibition appears essential for limiting oxidative metabolism linked to effector function in chronic T cells, thereby promoting survival and functional fitness.
Collapse
Affiliation(s)
- Marie Bettonville
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
| | - Stefania d'Aria
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
| | - Kathleen Weatherly
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
| | - Paolo E Porporato
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jinyu Zhang
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
| | - Sabrina Bousbata
- Laboratory of Molecular Parasitology, Proteomic Platform, Institute of Molecular Biology and Medicine, Université Libre de Bruxelles, Gosselies, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Michel Y Braun
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
| |
Collapse
|
68
|
Wong E, Davis JE, Grigg A, Szer J, Ritchie D. Strategies to enhance the graft versus tumour effect after allogeneic haematopoietic stem cell transplantation. Bone Marrow Transplant 2018; 54:175-189. [PMID: 29904127 DOI: 10.1038/s41409-018-0244-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 04/09/2018] [Accepted: 05/15/2018] [Indexed: 11/09/2022]
Abstract
Relapse of haematological malignancies after allogeneic haematopoietic stem cell transplant is a major cause of mortality. The immunological mechanisms that may lead to disease relapse may include immunological immaturity prior to reconstitution of the allogeneic immune system, tumour antigen downregulation or promotion of T-cell exhaustion by interactions with the tumour microenvironment. Current therapeutic strategies for post-transplant relapse are limited in their efficacy and alternative approaches are required. In this review, we discuss the mechanisms of T and NK-cell immune evasion that facilitate relapse of haematological malignancies after allogeneic stem cell transplantation, and explore emerging strategies to augment the allogeneic immune system in order to construct a more potent graft versus tumour response.
Collapse
Affiliation(s)
- Eric Wong
- Clinical Haematology and Bone Marrow Transplantation, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Victoria, Australia. .,Australian Cancer Research Foundation Translational Research Laboratory, Victoria, Australia. .,Department of Medicine, University of Melbourne, Victoria, Australia.
| | - Joanne E Davis
- Australian Cancer Research Foundation Translational Research Laboratory, Victoria, Australia.,Department of Medicine, University of Melbourne, Victoria, Australia
| | - Andrew Grigg
- Department of Medicine, University of Melbourne, Victoria, Australia.,Department of Clinical Haematology and Olivia Newton John Cancer Research Institute, Austin Hospital, Victoria, Australia
| | - Jeff Szer
- Clinical Haematology and Bone Marrow Transplantation, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Victoria, Australia.,Department of Medicine, University of Melbourne, Victoria, Australia
| | - David Ritchie
- Clinical Haematology and Bone Marrow Transplantation, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Victoria, Australia.,Australian Cancer Research Foundation Translational Research Laboratory, Victoria, Australia.,Department of Medicine, University of Melbourne, Victoria, Australia
| |
Collapse
|
69
|
Lamble AJ, Lind EF. Targeting the Immune Microenvironment in Acute Myeloid Leukemia: A Focus on T Cell Immunity. Front Oncol 2018; 8:213. [PMID: 29951373 PMCID: PMC6008423 DOI: 10.3389/fonc.2018.00213] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 05/24/2018] [Indexed: 12/27/2022] Open
Abstract
Immunotherapies, such as chimeric antigen receptor T cells, bispecific antibodies, and immune checkpoint inhibitors, have emerged as promising modalities in multiple hematologic malignancies. Despite the excitement surrounding immunotherapy, it is currently not possible to predict which patients will respond. Within solid tumors, the status of the immune microenvironment provides valuable insight regarding potential responses to immune therapies. Much less is known about the immune microenvironment within hematologic malignancies but the characteristics of this environment are likely to serve a similar predictive role. Acute myeloid leukemia (AML) is the most common hematologic malignancy in adults, and only 25% of patients are alive 5 years following their diagnosis. There is evidence that manipulation of the immune microenvironment by leukemia cells may play a role in promoting therapy resistance and disease relapse. In addition, it has long been documented that through modulation of the immune system following allogeneic bone marrow transplant, AML can be cured, even in patients with the highest risk disease. These concepts, along with the poor prognosis associated with this disease, have encouraged many groups to start exploring the utility of novel immune therapies in AML. While the implementation of these therapies into clinical trials for AML has been supported by preclinical rationale, many questions still exist surrounding their efficacy, tolerability, and the overall optimal approach. In this review, we discuss what is known about the immune microenvironment within AML with a specific focus on T cells and checkpoints, along with their implications for immune therapies.
Collapse
Affiliation(s)
- Adam J Lamble
- Pediatric Hematology/Oncology, Seattle Children's Hospital, Seattle, WA, United States
| | - Evan F Lind
- Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
70
|
Shindiapina P, Alinari L. Pembrolizumab and its role in relapsed/refractory classical Hodgkin's lymphoma: evidence to date and clinical utility. Ther Adv Hematol 2018; 9:89-105. [PMID: 29623180 PMCID: PMC5881987 DOI: 10.1177/2040620718761777] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 02/06/2018] [Indexed: 12/28/2022] Open
Abstract
Immune evasion is a critical mechanism of malignant cell survival, and relies in part on molecular signaling through the programmed cell death 1 (PD-1)/PD-1 ligand (PD-L1) axis that contributes to T cell exhaustion. Immune modulatory therapy with monoclonal antibodies against PD-1 designed to enhance antitumor immune response have shown promise in the treatment of advanced solid tumors and hematologic malignancies. Classical Hodgkin's lymphoma (cHL), a unique B cell malignancy characterized by an extensive but ineffective immune cell infiltrate surrounding a small number of tumor cells, has shown significant response to anti-PD-1 directed therapy. The anti-PD-1 monoclonal antibodies nivolumab and pembrolizumab have shown similarly remarkable activity in relapsed/refractory cHL and have been approved by the Food and Drug Administration for treatment of this disease. In this article we review the rationale of targeting the PD-1/PD-L1 axis in cHL and the pharmacology of pembrolizumab, and summarize the data on activity and safety profile of this agent in the treatment of relapsed/refractory cHL. We also discuss the potential benefits and pitfalls of using PD-1 blockade in the setting of allogeneic stem-cell transplantation, and summarize ongoing prospective trials of single-agent pembrolizumab and combination strategies as well as future directions.
Collapse
Affiliation(s)
- Polina Shindiapina
- Department of Internal Medicine, Division of Hematology, Arthur G James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Lapo Alinari
- Division of Hematology, Department of Internal Medicine, Ohio State University Wexner Medical Center, 410 West 12th Avenue, 481A Wiseman Hall, Columbus, Ohio, 43210, USA
| |
Collapse
|
71
|
Bair SM, Mato A, Svoboda J. Immunotherapy for the Treatment of Hodgkin Lymphoma: An Evolving Paradigm. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2018; 18:380-391. [PMID: 29685424 DOI: 10.1016/j.clml.2018.03.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/18/2018] [Accepted: 03/27/2018] [Indexed: 12/09/2022]
Abstract
Classical Hodgkin lymphoma (cHL) is one of the most common lymphomas in the Western world. Although most patients are cured with standard first-line therapy, up to 20% of patients will have relapsed or refractory disease. Although the conventional approach to treatment has consisted of chemotherapy, radiation, and for those who relapse, autologous or allogeneic transplantation, newer approaches have become available in recent years, including immunoconjugates and checkpoint inhibitors. These approaches have shown significant efficacy in clinical trials and might be associated with fewer long-term toxicities compared with conventional therapies. In this review we discuss the biology of cHL as it pertains to the immunosuppressive tumor microenvironment and then review the existing clinical trial results of several emerging immunotherapies in this context, including immune checkpoint inhibitors and adoptive cellular therapy. Finally, several clinical practice issues pertaining to the use of immunotherapies are discussed.
Collapse
Affiliation(s)
- Steven M Bair
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Anthony Mato
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jakub Svoboda
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA.
| |
Collapse
|
72
|
Cancer-associated oxidoreductase ERO1-α promotes immune escape through up-regulation of PD-L1 in human breast cancer. Oncotarget 2018; 8:24706-24718. [PMID: 28160557 PMCID: PMC5421881 DOI: 10.18632/oncotarget.14960] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 01/03/2017] [Indexed: 12/31/2022] Open
Abstract
Many human cancers have been reported to have enhanced expression of the immune checkpoint molecule programmed death-ligand 1 (PD-L1), which binds to programmed cell death-1 (PD-1) expressed on immune cells. PD-L1/PD-1 plays a role in inhibition of antitumor immunity by inducing T cell apoptosis and tolerance. Thus, it is crucial to elucidate mechanisms of PD-L1 expression on cancer cells. ERO1-α is an oxidase located in the endoplasmic reticulum. It is overexpressed in a variety of tumor types and it plays a role in disulfide bond formation in collaboration with PDI. Here, we investigated the influence of ERO1-α on expression of PD-L1 and immune escape. We demonstrated that ERO1-α augmented the expression of PD-L1 via facilitation of oxidative protein folding within PD-L1. In addition, we showed that overexpression of ERO1-α increased HIF-1α protein expression, resulting in an increase of PD-L1 mRNA as well as protein. In clinical cases, we observed that the expression of ERO1-α in triple negative breast cancer was related to the expression of PD-L1. Moreover, apoptosis of Jurkat leukemia T cells, which express PD-1, induced by tumor PD-L1 was inhibited when ERO1-α was depleted. The results suggest that targeting ERO1-α in tumor cells can be a novel approach for cancer immunotherapy. Therefore, the role of ERO1-α in tumor-mediated immunosuppression should be further explored.
Collapse
|
73
|
Abstract
Intricate systems of checkpoints such as the programmed cell death protein 1 (PD-1)/programmed death ligand 1 (PD-L1) axis regulate adaptive immune responses to protect against tissue damage. However, diverse cancers can exploit these pathways to evade or suppress antitumor immunity, leading to tumor progression. Correspondingly, immune checkpoint inhibitors that block PD-1/PD-L1 signaling have shown marked therapeutic efficacy in certain cancers, such as Hodgkin lymphoma. Reed-Sternberg cells, the hallmark cells of Hodgkin lymphoma, commonly overexpress PD-1 ligands, and recent clinical trials have demonstrated impressive response rates with the PD-1 inhibitors nivolumab and pembrolizumab in relapsed or refractory Hodgkin lymphoma, leading to their FDA approval in this setting. Current efforts are underway to improve clinical responses by incorporating PD-1 inhibitors into earlier treatment regimens and identifying therapeutic agents that synergize with PD-1 inhibitors. This review summarizes our understanding of the PD-1/PD-L1 axis in Hodgkin lymphoma, recent clinical studies of anti-PD-1 monotherapy and promising combination immunotherapy in the pipeline.
Collapse
|
74
|
Nguyen HD, Kuril S, Bastian D, Yu XZ. T-Cell Metabolism in Hematopoietic Cell Transplantation. Front Immunol 2018; 9:176. [PMID: 29479351 PMCID: PMC5811499 DOI: 10.3389/fimmu.2018.00176] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 01/19/2018] [Indexed: 12/11/2022] Open
Abstract
Metabolism, including catabolism and anabolism, is a basic cellular process necessary for cell survival. T lymphocytes have a distinct metabolism that can determine both fate and function. T-cell activation depends on glycolysis to obtain materials and energy for proliferation and effector function. Importantly, T cells utilize different metabolic processes under different conditions and diseases. Allogeneic hematopoietic cell transplantation (allo-HCT) is a classic immunotherapy for hematological malignancies; however, the development of graft-versus-host disease (GVHD) is a major factor limiting the success of allo-HCT. T cells in the donor graft drive GVHD by mounting a robust immunological attack against recipient normal tissues. Hence, understanding T-cell metabolism after allo-HCT would provide potential metabolic targets for the control of GVHD and primary tumor relapse. The purpose of the current review is to highlight the key metabolic pathways involved in alloantigen-activated T cells and to discuss how manipulating these pathways can serve as potential new therapeutic strategies to induce immune tolerance after allo-transplantation. We will also summarize the recent progress in regulating T-cell metabolism in bone marrow transplantation by targeting novel metabolic regulators or immune checkpoint molecules.
Collapse
Affiliation(s)
- Hung D Nguyen
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Sandeepkumar Kuril
- Department of Pediatric Ematology-Oncology, Medical University of South Carolina, Charleston, SC, United States
| | - David Bastian
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Xue-Zhong Yu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
75
|
Tyrosine kinase inhibitors and immune checkpoint blockade in allogeneic hematopoietic cell transplantation. Blood 2018; 131:1073-1080. [PMID: 29358177 DOI: 10.1182/blood-2017-10-752154] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 01/16/2018] [Indexed: 12/27/2022] Open
Abstract
Advances in the prevention of graft-versus-host disease (GVHD) and opportunistic infection have improved survival after allogeneic hematopoietic cell transplantation (allo-HCT) in the past decade. However, few inroads have been made into the treatment or prevention of relapse of the underlying malignancy for which allo-HCT is being performed. The introduction of US Food and Drug Administration-approved agents with significant activity in a variety of hematologic malignancies provides an opportunity to evaluate these interventions in the allo-HCT setting. Some of the most promising new agents include tyrosine kinase inhibitors (TKIs) directed at bcr-abl, kinase inhibitors targeting fms-like tyrosine kinase 3, and immune checkpoint inhibitors blocking both CTLA4 and PD-1. Data have emerged indicating potential efficacy of these agents in preventing or treating relapse, though definitive evidence remains elusive. However, potential toxicity can be considerable, highlighting the need for further clinical trials to define the therapeutic window. This review explores the immunologic and clinical consequence of treatment with both TKIs and checkpoint inhibitors in the peri- and post-allo-HCT setting.
Collapse
|
76
|
Chae YK, Galvez C, Anker JF, Iams WT, Bhave M. Cancer immunotherapy in a neglected population: The current use and future of T-cell-mediated checkpoint inhibitors in organ transplant patients. Cancer Treat Rev 2017; 63:116-121. [PMID: 29276997 DOI: 10.1016/j.ctrv.2017.12.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/04/2017] [Accepted: 12/06/2017] [Indexed: 12/22/2022]
Abstract
Although the indications for immune checkpoint inhibitors continue to grow, organ transplant recipients with advanced malignancies have been largely excluded from clinical trials testing the safety and efficacy of these therapies given their need for chronic immunosuppression and the risk of allograft rejection. With the rapid growth of transplant medicine and the increased risk of malignancy associated with chronic immunosuppression, it is critical that we systematically analyze the available data describing immune checkpoint blockade in the organ transplant population. Herein we provide a current and comprehensive review of cases in which immune checkpoint blockade was used on organ transplant recipients. Furthermore, we discuss the differences in efficacy and risk of allograft rejection between CTLA-4 and PD-1 inhibitors and make recommendations based on the limited available clinical data. We also discuss the future of immune checkpoint blockade in this subpopulation and explore the emerging data of promising combination therapies with mTOR, BRAF/MEK, and BTK/ITK inhibitors. Further clinical experience and larger clinical trials involving immune checkpoint inhibitors, whether as monotherapies or combinatorial therapies, will help develop regimens that optimize anti-tumor response and minimize the risk of allograft rejection in organ transplant patients.
Collapse
Affiliation(s)
- Young Kwang Chae
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.
| | - Carlos Galvez
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jonathan F Anker
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Wade T Iams
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Manali Bhave
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
77
|
Juchem KW, Sacirbegovic F, Zhang C, Sharpe AH, Russell K, McNiff JM, Demetris AJ, Shlomchik MJ, Shlomchik WD. PD-L1 Prevents the Development of Autoimmune Heart Disease in Graft-versus-Host Disease. THE JOURNAL OF IMMUNOLOGY 2017; 200:834-846. [PMID: 29212909 DOI: 10.4049/jimmunol.1701076] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 11/03/2017] [Indexed: 01/22/2023]
Abstract
Effector memory T cells (TEM) are less capable of inducing graft-versus-host disease (GVHD) compared with naive T cells (TN). Previously, in the TS1 TCR transgenic model of GVHD, wherein TS1 CD4 cells specific for a model minor histocompatibility Ag (miHA) induce GVHD in miHA-positive recipients, we found that cell-intrinsic properties of TS1 TEM reduced their GVHD potency relative to TS1 TN Posttransplant, TS1 TEM progeny expressed higher levels of PD-1 than did TS1 TN progeny, leading us to test the hypothesis that TEM induce less GVHD because of increased sensitivity to PD-ligands. In this study, we tested this hypothesis and found that indeed TS1 TEM induced more severe skin and liver GVHD in the absence of PD-ligands. However, lack of PD-ligands did not result in early weight loss and colon GVHD comparable to that induced by TS1 TN, indicating that additional pathways restrain alloreactive TEM TS1 TN also caused more severe GVHD without PD-ligands. The absence of PD-ligands on donor bone marrow was sufficient to augment GVHD caused by either TEM or TN, indicating that donor PD-ligand-expressing APCs critically regulate GVHD. In the absence of PD-ligands, both TS1 TEM and TN induced late-onset myocarditis. Surprisingly, this was an autoimmune manifestation, because its development required non-TS1 polyclonal CD8+ T cells. Myocarditis development also required donor bone marrow to be PD-ligand deficient, demonstrating the importance of donor APC regulatory function. In summary, PD-ligands suppress both miHA-directed GVHD and the development of alloimmunity-induced autoimmunity after allogeneic hematopoietic transplantation.
Collapse
Affiliation(s)
- Kathryn W Juchem
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | | | - Cuiling Zhang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Arlene H Sharpe
- Department of Microbiology and Immunology, Harvard Medical School, Boston, MA 02115
| | - Kerry Russell
- Department of Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Jennifer M McNiff
- Department of Dermatology, Yale University School of Medicine, New Haven, CT 06520.,Department of Pathology, Yale University School of Medicine, New Haven, CT 06520
| | | | - Mark J Shlomchik
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520.,Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520; and.,Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261
| | - Warren D Shlomchik
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520; .,Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261.,Department of Medicine, Yale University School of Medicine, New Haven, CT 06520.,Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261
| |
Collapse
|
78
|
Xu-Monette ZY, Zhang M, Li J, Young KH. PD-1/PD-L1 Blockade: Have We Found the Key to Unleash the Antitumor Immune Response? Front Immunol 2017; 8:1597. [PMID: 29255458 PMCID: PMC5723106 DOI: 10.3389/fimmu.2017.01597] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 11/06/2017] [Indexed: 12/13/2022] Open
Abstract
PD-1-PD-L1 interaction is known to drive T cell dysfunction, which can be blocked by anti-PD-1/PD-L1 antibodies. However, studies have also shown that the function of the PD-1-PD-L1 axis is affected by the complex immunologic regulation network, and some CD8+ T cells can enter an irreversible dysfunctional state that cannot be rescued by PD-1/PD-L1 blockade. In most advanced cancers, except Hodgkin lymphoma (which has high PD-L1/L2 expression) and melanoma (which has high tumor mutational burden), the objective response rate with anti-PD-1/PD-L1 monotherapy is only ~20%, and immune-related toxicities and hyperprogression can occur in a small subset of patients during PD-1/PD-L1 blockade therapy. The lack of efficacy in up to 80% of patients was not necessarily associated with negative PD-1 and PD-L1 expression, suggesting that the roles of PD-1/PD-L1 in immune suppression and the mechanisms of action of antibodies remain to be better defined. In addition, important immune regulatory mechanisms within or outside of the PD-1/PD-L1 network need to be discovered and targeted to increase the response rate and to reduce the toxicities of immune checkpoint blockade therapies. This paper reviews the major functional and clinical studies of PD-1/PD-L1, including those with discrepancies in the pathologic and biomarker role of PD-1 and PD-L1 and the effectiveness of PD-1/PD-L1 blockade. The goal is to improve understanding of the efficacy of PD-1/PD-L1 blockade immunotherapy, as well as enhance the development of therapeutic strategies to overcome the resistance mechanisms and unleash the antitumor immune response to combat cancer.
Collapse
Affiliation(s)
- Zijun Y. Xu-Monette
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jianyong Li
- Department of Hematology, JiangSu Province Hospital, The First Affiliated Hospital of NanJing Medical University, NanJing, JiangSu Province, China
| | - Ken H. Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Graduate School of Biomedical Science, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
79
|
Affiliation(s)
- Robert Zeiser
- From the Department of Hematology, Oncology, and Stem Cell Transplantation, Faculty of Medicine, Freiburg University Medical Center, Freiburg, Germany (R.Z.); and the Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis (B.R.B.)
| | - Bruce R Blazar
- From the Department of Hematology, Oncology, and Stem Cell Transplantation, Faculty of Medicine, Freiburg University Medical Center, Freiburg, Germany (R.Z.); and the Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis (B.R.B.)
| |
Collapse
|
80
|
Lucchesi M, Sardi I, Puppo G, Chella A, Favre C. The dawn of "immune-revolution" in children: early experiences with checkpoint inhibitors in childhood malignancies. Cancer Chemother Pharmacol 2017; 80:1047-1053. [PMID: 29067473 DOI: 10.1007/s00280-017-3450-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 10/09/2017] [Indexed: 10/18/2022]
Abstract
Modern immunotherapy with checkpoint inhibitors has changed clinical practice of adult patients with advanced cancer. Blockade of CTLA-4 and PD-1 pathways have shown survival benefits in different diseases. In children, combination of surgery, radiotherapy and chemotherapy have improved survival rates of solid tumors. However, the outcomes for subsets of patients such as those with high-grade, refractory, or metastatic disease remain extremely poor. Currently, the treatment of these patients is almost exclusively based on standard chemotherapy. The significant proportion of pediatric cancers with high number of mutations and subsequent high expression of neoantigens, together with the potential prognostic role of the immunosuppressive checkpoint molecules (CTLA-4, PD-L1) can represent a promising rationale that support the use of checkpoint inhibitors. We made a revision about emerging data regarding safety and activity of checkpoint inhibitors in children with solid tumors.
Collapse
Affiliation(s)
- Maurizio Lucchesi
- Pulmonology Unit, Thoracic Cancer Center, Azienda Ospedaliero-Universitaria Pisana, Via Roma, 67, 56126, Pisa, Italy. .,Tuscany Network for Paediatric Oncology-Istituto Toscano Tumori (ITT), Florence, Italy.
| | - Iacopo Sardi
- Neuro-Oncology Unit, Anna Meyer Children's Hospital, Florence, Italy.,Department of Pediatric Oncology, Hematology, and Transplants, Anna Meyer Children's Hospital, Florence, Italy.,Tuscany Network for Paediatric Oncology-Istituto Toscano Tumori (ITT), Florence, Italy
| | - Gianfranco Puppo
- Pulmonology Unit, Thoracic Cancer Center, Azienda Ospedaliero-Universitaria Pisana, Via Roma, 67, 56126, Pisa, Italy
| | - Antonio Chella
- Pulmonology Unit, Thoracic Cancer Center, Azienda Ospedaliero-Universitaria Pisana, Via Roma, 67, 56126, Pisa, Italy.,Tuscany Network for Paediatric Oncology-Istituto Toscano Tumori (ITT), Florence, Italy
| | - Claudio Favre
- Department of Pediatric Oncology, Hematology, and Transplants, Anna Meyer Children's Hospital, Florence, Italy.,Tuscany Network for Paediatric Oncology-Istituto Toscano Tumori (ITT), Florence, Italy
| |
Collapse
|
81
|
Davoodzadeh Gholami M, Kardar GA, Saeedi Y, Heydari S, Garssen J, Falak R. Exhaustion of T lymphocytes in the tumor microenvironment: Significance and effective mechanisms. Cell Immunol 2017; 322:1-14. [PMID: 29079339 DOI: 10.1016/j.cellimm.2017.10.002] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 10/08/2017] [Accepted: 10/09/2017] [Indexed: 12/23/2022]
Abstract
T lymphocytes play crucial roles in adaptive immune responses to tumors. However, due to different tolerance mechanisms and inhibitory effects of the tumor microenvironment (TME) on T cells, responses to tumors are insufficient. In fact, cellular and molecular suppressive mechanisms repress T cell responses in the TME, resulting in senescent, anergic and exhausted lymphocytes. Exhaustion is a poor responsive status of T cells, with up-regulated expression of inhibitory receptors, decreased production of effective cytokines, and reduced cytotoxic activity. Low immunogenicity of tumor antigens and inadequate presentation of tumor-specific antigens results in inappropriate activation of naive T lymphocytes against tumor antigens. Moreover, when effector cytotoxic T cells enter TME, they encounter a complicated network of cells and cytokines that suppress their effectiveness and turn them into exhausted T cells. Thus, the mechanism of T cell exhaustion in cancer is different from that in chronic infections. In this review we will discuss the main components such as inhibitory receptors, inflammatory cells, stromal cells, cytokine milieu as well as environmental and metabolic conditions in TME which play role in development of exhaustion. Furthermore, recent therapeutic methods available to overcome exhaustion will be discussed.
Collapse
Affiliation(s)
- Mohammad Davoodzadeh Gholami
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Gholam Ali Kardar
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Yousef Saeedi
- Department of Pharmaceutical Sciences, Utrecht University, Netherlands.
| | - Sahel Heydari
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Johan Garssen
- Department of Pharmaceutical Sciences, Utrecht University, Netherlands.
| | - Reza Falak
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
82
|
Hossain MS, Kunter GM, El-Najjar VF, Jaye DL, Al-Kadhimi Z, Taofeek OK, Li JM, Waller EK. PD-1 and CTLA-4 up regulation on donor T cells is insufficient to prevent GvHD in allo-HSCT recipients. PLoS One 2017; 12:e0184254. [PMID: 28953925 PMCID: PMC5617147 DOI: 10.1371/journal.pone.0184254] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 08/21/2017] [Indexed: 01/22/2023] Open
Abstract
The expression of checkpoint blockade molecules PD-1, PD-L1, CTLA-4, and foxp3+CD25+CD4+ T cells (Tregs) regulate donor T cell activation and graft-vs-host disease (GvHD) in allogeneic hematopoietic stem cell transplant (allo-HSCT). Detailed kinetics of PD-1-, CTLA-4-, and PD-L1 expression on donor and host cells in GvHD target organs have not been well studied. Using an established GvHD model of allo-HSCT (B6 → CB6F1), we noted transient increases of PD-1- and CTLA-4-expressing donor CD4+ and CD8+ T cells on day 10 post transplant in spleens of allo-HSCT recipients compared with syngeneic HSCT (syn-HSCT) recipients. In contrast, expression of PD-1- and CTLA-4 on donor T cells was persistently increased in bone marrow (BM) of allo-HSCT recipients compared with syn-HSCT recipients. Similar differential patterns of donor T cell immune response were observed in a minor histocompatibility (miHA) mismatched transplant model of GvHD. Despite higher PD-1 and CTLA-4 expression in BM, numbers of foxp3+ T cells and Tregs were much lower in allo-HSCT recipients compared with syn-HSCT recipients. PD-L1-expressing host cells were markedly decreased concomitant with elimination of residual host hematopoietic elements in spleens of allo-HSCT recipients. Allo-HSCT recipients lacking PD-L1 rapidly developed increased serum inflammatory cytokines and lethal acute GvHD compared with wild-type (WT) B6 allo-HSCT recipients. These data suggest that increased expression of checkpoint blockade molecules PD-1 and CTLA-4 on donor T cells is not sufficient to prevent GvHD, and that cooperation between checkpoint blockade signaling by host cells and donor Tregs is necessary to limit GvHD in allo-HSCT recipients.
Collapse
Affiliation(s)
- Mohammad S. Hossain
- Department of Hematology and Medical Oncology, Division of Stem Cell and Bone Marrow Transplantation, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Ghada M. Kunter
- Holzer Cancer Center, Gallipolis, Ohio, United States of America
| | - Vicky F. El-Najjar
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - David L. Jaye
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Zaid Al-Kadhimi
- Department of Hematology and Medical Oncology, Division of Stem Cell and Bone Marrow Transplantation, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Owonikoko K. Taofeek
- Department of Hematology and Medical Oncology, Division of Stem Cell and Bone Marrow Transplantation, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Jian-Ming Li
- Department of Hematology and Medical Oncology, Division of Stem Cell and Bone Marrow Transplantation, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Edmund K. Waller
- Department of Hematology and Medical Oncology, Division of Stem Cell and Bone Marrow Transplantation, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
83
|
Merryman RW, Armand P. Immune Checkpoint Blockade and Hematopoietic Stem Cell Transplant. Curr Hematol Malig Rep 2017; 12:44-50. [PMID: 28155012 DOI: 10.1007/s11899-017-0362-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Allogeneic hematopoietic stem cell transplant (HSCT) relies primarily upon graft-versus-tumor activity for cancer eradication. Relapse remains the principal cause of treatment failure after HSCT, implying frequent immune escape, which in at least some cases, appears to be mediated by increased expression of inhibitory immune checkpoints. In an attempt to restore anti-tumor immunity, checkpoint blockade therapy (CBT) targeting PD-1 and CLTA-4 has been used in conjunction with both allogeneic and autologous HSCT. Clinical experience in this setting is limited to several small clinical trials and case series, but together they suggest that treatment with CBT can effectively amplify anti-tumor immune responses. However, intrinsic to its mechanism is also the risk that CBT in the HSCT setting may also cause significant immune toxicity. Fatal immune-related adverse events and graft-versus-host disease have been observed, but in most cases, immune side effects appear to be reversible with steroids and CBT discontinuation. As clinical investigation continues, improved understanding of immune checkpoint biology will be critical to optimize safe and efficacious treatment strategies.
Collapse
Affiliation(s)
- Reid W Merryman
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Philippe Armand
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA.
| |
Collapse
|
84
|
Gibbons Johnson RM, Dong H. Functional Expression of Programmed Death-Ligand 1 (B7-H1) by Immune Cells and Tumor Cells. Front Immunol 2017; 8:961. [PMID: 28848559 PMCID: PMC5554355 DOI: 10.3389/fimmu.2017.00961] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 07/27/2017] [Indexed: 01/05/2023] Open
Abstract
The programmed death-1 (PD-1) and its ligand PD-L1 (B7-H1) signaling pathway has been the focus of much enthusiasm in the fields of tumor immunology and oncology with recent FDA approval of the anti-PD-1 antibodies pembrolizumab and nivolumab and the anti-PD-L1 antibodies durvalumab, atezolimuab, and avelumab. These therapies, referred to here as PD-L1/PD-1 checkpoint blockade therapies, are designed to block the interaction between PD-L1, expressed by tumor cells, and PD-1, expressed by tumor-infiltrating CD8+ T cells, leading to enhanced antitumor CD8+ T cell responses and tumor regression. The influence of PD-L1 expressed by tumor cells on antitumor CD8+ T cell responses is well characterized, but the impact of PD-L1 expressed by immune cells has not been well defined for antitumor CD8+ T cell responses. Although PD-L1 expression by tumor cells has been used as a biomarker in selection of patients for PD-L1/PD-1 checkpoint blockade therapies, patients whose tumor cells lack PD-L1 expression often respond positively to PD-L1/PD-1 checkpoint blockade therapies. This suggests that PD-L1 expressed by non-malignant cells may also contribute to antitumor immunity. Here, we review the functions of PD-L1 expressed by immune cells in the context of CD8+ T cell priming, contraction, and differentiation into memory populations, as well as the role of PD-L1 expressed by tumor cells in regulating antitumor CD8+ T cell responses.
Collapse
Affiliation(s)
| | - Haidong Dong
- Department of Urology, College of Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Immunology, College of Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
85
|
Association between GvHD and nivolumab in the FDA adverse event reporting system. Bone Marrow Transplant 2017; 52:1463-1464. [PMID: 28759026 DOI: 10.1038/bmt.2017.158] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
86
|
Boddu P, Kantarjian H, Garcia-Manero G, Allison J, Sharma P, Daver N. The emerging role of immune checkpoint based approaches in AML and MDS. Leuk Lymphoma 2017; 59:790-802. [PMID: 28679300 DOI: 10.1080/10428194.2017.1344905] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The development of immune checkpoint inhibitors represents a major breakthrough in the field of cancer therapeutics. Pursuant to their success in melanoma and numerous solid tumor malignancies, these agents are being investigated in hematological malignancies including acute myelogenous leukemia (AML) and myelodysplastic syndromes (MDS). Although AML/MDS have traditionally been considered to be less immunogenic than solid tumor malignancies, recent pre-clinical models suggest a therapeutic role for immune checkpoint inhibition in these diseases. CTLA-4 inhibition may be especially effective in treating late post-allogeneic stem cell transplant relapse of AML in patients with limited or no graft versus host disease. Immune checkpoint inhibition, specifically PD-1 inhibition, demonstrated limited single agent efficacy in patients with relapsed AML and with MDS post-hypomethylating therapy. Rationally designed combinations of PD-1 inhibitors with standard anti-leukemic therapy are needed. Hypomethylating agents such as azacitidine, up-regulate PD-1, PD-L1, and PD-L2 in patients with AML/MDS and up-regulation of these genes was associated with the emergence of resistance. The combination of azacitidine and PD-1/PD-L1 inhibition may be a potential mechanism to prevent or overcome resistance to 5-azacitidine. A number of such combinations are being evaluated in clinical trials with early encouraging results. Immune checkpoint inhibition is also an attractive option to improve relapse-free survival or eliminate minimal residual disease post induction and consolidation by enhancing T-cell surveillance in patients with high-risk AML. The ongoing clinical trials with checkpoint inhibitors in AML/MDS will improve our understanding of the immunobiology of these diseases and guide us to the most appropriate application of these agents in the therapy of AML/MDS.
Collapse
Affiliation(s)
- Prajwal Boddu
- a Department of Leukemia , The University of Texas M. D. Anderson Cancer Center , Houston , TX , USA
| | - Hagop Kantarjian
- a Department of Leukemia , The University of Texas M. D. Anderson Cancer Center , Houston , TX , USA
| | - Guillermo Garcia-Manero
- a Department of Leukemia , The University of Texas M. D. Anderson Cancer Center , Houston , TX , USA
| | - James Allison
- a Department of Leukemia , The University of Texas M. D. Anderson Cancer Center , Houston , TX , USA
| | - Padmanee Sharma
- b Immunotherapy Platform , The University of Texas M. D. Anderson Cancer Center , Houston , TX , USA
| | - Naval Daver
- a Department of Leukemia , The University of Texas M. D. Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
87
|
Dunavin N, Dias A, Li M, McGuirk J. Mesenchymal Stromal Cells: What Is the Mechanism in Acute Graft-Versus-Host Disease? Biomedicines 2017; 5:biomedicines5030039. [PMID: 28671556 PMCID: PMC5618297 DOI: 10.3390/biomedicines5030039] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 06/06/2017] [Accepted: 06/14/2017] [Indexed: 12/23/2022] Open
Abstract
After more than a decade of preclinical and clinical development, therapeutic infusion of mesenchymal stromal cells is now a leading investigational strategy for the treatment of acute graft-versus-host disease (GVHD). While their clinical use continues to expand, it is still unknown which of their immunomodulatory properties contributes most to their therapeutic activity. Herein we describe the proposed mechanisms, focusing on the inhibitory activity of mesenchymal stromal cells (MSCs) at immunologic checkpoints. A deeper understanding of the mechanism of action will allow us to design more effective treatment strategies.
Collapse
Affiliation(s)
- Neil Dunavin
- Division of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Cancer Center, 2330 Shawnee Mission Pkwy., Westwood, KS 66205, USA.
| | - Ajoy Dias
- Division of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Cancer Center, 2330 Shawnee Mission Pkwy., Westwood, KS 66205, USA.
| | - Meizhang Li
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA.
| | - Joseph McGuirk
- Division of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Cancer Center, 2330 Shawnee Mission Pkwy., Westwood, KS 66205, USA.
| |
Collapse
|
88
|
Leigh ND, O'Neill RE, Du W, Chen C, Qiu J, Ashwell JD, McCarthy PL, Chen GL, Cao X. Host-Derived CD70 Suppresses Murine Graft-versus-Host Disease by Limiting Donor T Cell Expansion and Effector Function. THE JOURNAL OF IMMUNOLOGY 2017; 199:336-347. [PMID: 28550198 DOI: 10.4049/jimmunol.1502181] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 05/01/2017] [Indexed: 11/19/2022]
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is a potentially curative treatment for hematologic and immunologic diseases. However, graft-versus-host disease (GVHD) may develop when donor-derived T cells recognize and damage genetically distinct normal host tissues. In addition to TCR signaling, costimulatory pathways are involved in T cell activation. CD27 is a TNFR family member expressed on T cells, and its ligand, CD70, is expressed on APCs. The CD27/CD70 costimulatory pathway was shown to be critical for T cell function and survival in viral infection models. However, the role of this pathway in allo-HCT is previously unknown. In this study, we have examined its contribution in GVHD pathogenesis. Surprisingly, Ab blockade of CD70 after allo-HCT significantly increases GVHD. Interestingly, whereas donor T cell- or bone marrow-derived CD70 plays no role in GVHD, host-derived CD70 inhibits GVHD as CD70-/- hosts show significantly increased GVHD. This is evidenced by reduced survival, more severe weight loss, and increased histopathologic damage compared with wild-type hosts. In addition, CD70-/- hosts have higher levels of proinflammatory cytokines TNF-α, IFN-γ, IL-2, and IL-17. Moreover, accumulation of donor CD4+ and CD8+ effector T cells is increased in CD70-/- versus wild-type hosts. Mechanistic analyses suggest that CD70 expressed by host hematopoietic cells is involved in the control of alloreactive T cell apoptosis and expansion. Together, our findings demonstrate that host CD70 serves as a unique negative regulator of allogeneic T cell response by contributing to donor T cell apoptosis and inhibiting expansion of donor effector T cells.
Collapse
Affiliation(s)
- Nicholas D Leigh
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263
| | - Rachel E O'Neill
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263
| | - Wei Du
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263
| | - Chuan Chen
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263
| | - Jingxin Qiu
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY 14263
| | - Jonathan D Ashwell
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Philip L McCarthy
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY 14263
| | - George L Chen
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY 14263
| | - Xuefang Cao
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263;
| |
Collapse
|
89
|
Bond DA, Alinari L. Emerging treatment options for the management of Hodgkin's lymphoma: clinical utility of nivolumab. J Blood Med 2017; 8:41-54. [PMID: 28546779 PMCID: PMC5436782 DOI: 10.2147/jbm.s117452] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Classical Hodgkin's lymphoma (cHL) is a B-cell malignancy comprised of pathologic Reed Sternberg cells with a surrounding immune-tolerant inflammatory milieu. RS cells evade immune recognition in part through programmed death ligand 1 (PD-L1) overexpression, which is genetically programmed through copy number alterations, polysomy, and amplification of the 9p24.1 locus encoding PD-L1. By engaging with PD-1+ T-cells, PD-L1 delivers a potent immune suppressive signal promoting immunologic escape of the tumor cell. Enhancing antitumor immune response by targeting PD-1 with the monoclonal antibody nivolumab has proved to be effective in multiple solid tumors, but the highest response rates to date have been reported in patients with cHL, with over 65% of treated patients achieving an objective clinical response. In this review, we will summarize the published evidence regarding the activity of nivolumab in cHL as well as its current place in therapy. We will review the pharmacology, mechanism of action, and side effects of nivolumab as well as the emerging data indicating possible increased risk of graft versus host disease in patients treated with PD-1 inhibitors either pre- or post-allogeneic stem cell transplant. Given the remarkable single-agent activity and safety profile of PD-1 inhibitors in heavily pretreated patients with cHL, the possibility of employing nivolumab in combination with other active agents and earlier in therapy is a promising area of active investigation, and we will briefly summarize current clinical trials.
Collapse
Affiliation(s)
- David A Bond
- Department of Internal Medicine, Division of Hematology, Arthur G James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Lapo Alinari
- Department of Internal Medicine, Division of Hematology, Arthur G James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
90
|
PD-1 blockade for relapsed lymphoma post-allogeneic hematopoietic cell transplant: high response rate but frequent GVHD. Blood 2017; 130:221-228. [PMID: 28468799 DOI: 10.1182/blood-2017-01-761346] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 04/24/2017] [Indexed: 12/21/2022] Open
Abstract
Given the limited treatment options for relapsed lymphoma post-allogeneic hematopoietic cell transplantation (post-allo-HCT) and the success of programmed death 1 (PD-1) blockade in classical Hodgkin lymphoma (cHL) patients, anti-PD-1 monoclonal antibodies (mAbs) are increasingly being used off-label after allo-HCT. To characterize the safety and efficacy of PD-1 blockade in this setting, we conducted a multicenter retrospective analysis of 31 lymphoma patients receiving anti-PD-1 mAbs for relapse post-allo-HCT. Twenty-nine (94%) patients had cHL and 27 had ≥1 salvage therapy post-allo-HCT and prior to anti-PD-1 treatment. Median follow-up was 428 days (range, 133-833) after the first dose of anti-PD-1. Overall response rate was 77% (15 complete responses and 8 partial responses) in 30 evaluable patients. At last follow-up, 11 of 31 patients progressed and 21 of 31 (68%) remain alive, with 8 (26%) deaths related to new-onset graft-versus-host disease (GVHD) after anti-PD-1. Seventeen (55%) patients developed treatment-emergent GVHD after initiation of anti-PD-1 (6 acute, 4 overlap, and 7 chronic), with onset after a median of 1, 2, and 2 doses, respectively. GVHD severity was grade III-IV acute or severe chronic in 9 patients. Only 2 of these 17 patients achieved complete response to GVHD treatment, and 14 of 17 required ≥2 systemic therapies. In conclusion, PD-1 blockade in relapsed cHL allo-HCT patients appears to be highly efficacious but frequently complicated by rapid onset of severe and treatment-refractory GVHD. PD-1 blockade post-allo-HCT should be studied further but cannot be recommended for routine use outside of a clinical trial.
Collapse
|
91
|
Kasamon YL, de Claro RA, Wang Y, Shen YL, Farrell AT, Pazdur R. FDA Approval Summary: Nivolumab for the Treatment of Relapsed or Progressive Classical Hodgkin Lymphoma. Oncologist 2017; 22:585-591. [PMID: 28438889 PMCID: PMC5423515 DOI: 10.1634/theoncologist.2017-0004] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 02/08/2017] [Indexed: 11/17/2022] Open
Abstract
On May 17, 2016, after an expedited priority review, the U.S. Food and Drug Administration granted accelerated approval to nivolumab for the treatment of patients with classical Hodgkin lymphoma (cHL) that has relapsed or progressed after autologous hematopoietic stem cell transplantation (HSCT) and post-transplantation brentuximab vedotin (BV). Nivolumab in cHL had been granted breakthrough therapy designation. Accelerated approval was based on two single-arm, multicenter trials in adults with cHL. In 95 patients with relapsed or progressive cHL after autologous HSCT and post-transplantation BV, nivolumab, dosed at 3 mg/kg intravenously every 2 weeks, produced a 65% (95% confidence interval: 55%-75%) objective response rate (58% partial remission, 7% complete remission). The estimated median duration of response was 8.7 months, with 4.6-month median follow-up for response duration. The median time to response was 2.1 (range: 0.7-5.7) months. Among 263 patients with cHL treated with nivolumab, 21% reported serious adverse reactions (ARs). The most common all-grade ARs (reported in ≥20%) were fatigue, upper respiratory tract infection, cough, pyrexia, diarrhea, elevated transaminases, and cytopenias. Infusion-related reaction and hypothyroidism or thyroiditis occurred in >10% of patients; other immune-mediated ARs, occurring in 1%-5%, included rash, pneumonitis, hepatitis, hyperthyroidism, and colitis. A new Warning and Precaution was issued for complications of allogeneic HSCT after nivolumab, including severe or hyperacute graft-versus-host disease, other immune-mediated ARs, and transplant-related mortality. Continued approval for the cHL indication may be contingent upon verification of clinical benefit in a randomized trial. The Oncologist 2017;22:585-591 IMPLICATIONS FOR PRACTICE: Based on response rate and duration in single-arm studies, nivolumab is a new treatment option for patients with classical Hodgkin lymphoma (cHL) that has relapsed or progressed despite autologous hematopoietic stem cell transplantation (HSCT) and brentuximab vedotin. This was the first U.S. Food and Drug Administration marketing application for a programmed cell death 1 inhibitor in hematologic malignancies. The use of immune checkpoint blockade in cHL represents a new treatment paradigm. The safety of allogeneic HSCT after nivolumab requires further evaluation, as does the safety of nivolumab after allogeneic HSCT.
Collapse
Affiliation(s)
- Yvette L Kasamon
- Office of Hematology and Oncology Products and Office of Biostatistics, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - R Angelo de Claro
- Office of Hematology and Oncology Products and Office of Biostatistics, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yaping Wang
- Office of Hematology and Oncology Products and Office of Biostatistics, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yuan Li Shen
- Office of Hematology and Oncology Products and Office of Biostatistics, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ann T Farrell
- Office of Hematology and Oncology Products and Office of Biostatistics, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Richard Pazdur
- Office of Hematology and Oncology Products and Office of Biostatistics, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
92
|
Beckermann KE, Dudzinski SO, Rathmell JC. Dysfunctional T cell metabolism in the tumor microenvironment. Cytokine Growth Factor Rev 2017; 35:7-14. [PMID: 28456467 DOI: 10.1016/j.cytogfr.2017.04.003] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 04/19/2017] [Indexed: 12/17/2022]
Abstract
Metabolic and signaling pathways are integrated to determine T cell fate and function. As stimulated T cells gain distinct effector functions, specific metabolic programs and demands are also adopted. These changes are essential for T cell effector function, and alterations or dysregulation of metabolic pathways can modulate T cell function. One physiological setting that impacts T cell metabolism is the tumor microenvironment. The metabolism of cancer cells themselves can limit nutrients and accumulate waste products. In addition to the expression of inhibitory ligands that directly modify T cell physiology, T cell metabolism may be strongly inhibited in the tumor microenvironment. This suppression of T cell metabolism may inhibit effector T cell activity while promoting suppressive regulatory T cells, and act as a barrier to effective immunotherapies. A thorough understanding of the effect of the tumor microenvironment on the immune system will support the continued improvement of immune based therapies for cancer patients.
Collapse
Affiliation(s)
- Kathryn E Beckermann
- Department of Medicine, Division of Hematology/Oncology, Vanderbilt-Ingram Cancer Center, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Stephanie O Dudzinski
- Department of Biomedical Engineering, 2301 Vanderbilt Place, Nashville, TN 37235, USA
| | - Jeffrey C Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 1211 Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
93
|
Ni X, Song Q, Cassady K, Deng R, Jin H, Zhang M, Dong H, Forman S, Martin PJ, Chen YZ, Wang J, Zeng D. PD-L1 interacts with CD80 to regulate graft-versus-leukemia activity of donor CD8+ T cells. J Clin Invest 2017; 127:1960-1977. [PMID: 28414296 PMCID: PMC5409099 DOI: 10.1172/jci91138] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 01/31/2017] [Indexed: 12/16/2022] Open
Abstract
Programmed death ligand-1 (PD-L1) interacts with programmed death-1 (PD-1) and the immunostimulatory molecule CD80 and functions as a checkpoint to regulate immune responses. The interaction of PD-L1 with CD80 alone has been shown to exacerbate the severity of graft-versus-host disease (GVHD), whereas costimulation of CD80 and PD-1 ameliorates GVHD. Here we have demonstrated that temporary depletion of donor CD4+ T cells early after hematopoietic cell transplantation effectively prevents GVHD while preserving strong graft-versus-leukemia (GVL) effects in allogeneic and xenogeneic murine GVHD models. Depletion of donor CD4+ T cells increased serum IFN-γ but reduced IL-2 concentrations, leading to upregulation of PD-L1 expression by recipient tissues and donor CD8+ T cells. In GVHD target tissues, the interactions of PD-L1 with PD-1 on donor CD8+ T cells cause anergy, exhaustion, and apoptosis, thereby preventing GVHD. In lymphoid tissues, the interactions of PD-L1 with CD80 augment CD8+ T cell expansion without increasing anergy, exhaustion, or apoptosis, resulting in strong GVL effects. These results indicate that the outcome of PD-L1-mediated signaling in CD8+ T cells depends on the presence or absence of CD4+ T cells, the nature of the interacting receptor expressed by CD8+ T cells, and the tissue environment in which the signaling occurs.
Collapse
|
94
|
Gupta S, Roy A, Dwarakanath BS. Metabolic Cooperation and Competition in the Tumor Microenvironment: Implications for Therapy. Front Oncol 2017; 7:68. [PMID: 28447025 PMCID: PMC5388702 DOI: 10.3389/fonc.2017.00068] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 03/24/2017] [Indexed: 12/31/2022] Open
Abstract
The tumor microenvironment (TME) is an ensemble of non-tumor cells comprising fibroblasts, cells of the immune system, and endothelial cells, besides various soluble secretory factors from all cellular components (including tumor cells). The TME forms a pro-tumorigenic cocoon around the tumor cells where reprogramming of the metabolism occurs in tumor and non-tumor cells that underlies the nature of interactions as well as competitions ensuring steady supply of nutrients and anapleoretic molecules for the tumor cells that fuels its growth even under hypoxic conditions. This metabolic reprogramming also plays a significant role in suppressing the immune attack on the tumor cells and in resistance to therapies. Thus, the metabolic cooperation and competition among the different TME components besides the inherent alterations in the tumor cells arising out of genetic as well as epigenetic changes supports growth, metastasis, and therapeutic resistance. This review focuses on the metabolic remodeling achieved through an active cooperation and competition among the three principal components of the TME—the tumor cells, the T cells, and the cancer-associated fibroblasts while discussing about the current strategies that target metabolism of TME components. Further, we will also consider the probable therapeutic opportunities targeting the various metabolic pathways as well as the signaling molecules/transcription factors regulating them for the development of novel treatment strategies for cancer.
Collapse
Affiliation(s)
- Seema Gupta
- Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Amrita Roy
- School of Life Sciences, B. S. Abdur Rahman Crescent University, Chennai, India
| | | |
Collapse
|
95
|
Patsoukis N, Weaver JD, Strauss L, Herbel C, Seth P, Boussiotis VA. Immunometabolic Regulations Mediated by Coinhibitory Receptors and Their Impact on T Cell Immune Responses. Front Immunol 2017; 8:330. [PMID: 28443090 PMCID: PMC5387055 DOI: 10.3389/fimmu.2017.00330] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 03/08/2017] [Indexed: 12/18/2022] Open
Abstract
Host immunity provides wide spectrum protection that serves to eradicate pathogens and cancer cells, while maintaining self-tolerance and immunological homeostasis. Ligation of the T cell receptor (TCR) by antigen activates signaling pathways that coordinately induce aerobic glycolysis, mitochondrial activity, anabolic metabolism, and T effector cell differentiation. Activation of PI3K, Akt, and mTOR triggers the switch to anabolic metabolism by inducing transcription factors such as Myc and HIF1, and the glucose transporter Glut1, which is pivotal for the increase of glucose uptake after T cell activation. Activation of MAPK signaling is required for glucose and glutamine utilization, whereas activation of AMPK is critical for energy balance and metabolic fitness of T effector and memory cells. Coinhibitory receptors target TCR-proximal signaling and generation of second messengers. Imbalanced activation of such signaling pathways leads to diminished rates of aerobic glycolysis and impaired mitochondrial function resulting in defective anabolic metabolism and altered T cell differentiation. The coinhibitory receptors mediate distinct and synergistic effects on the activation of signaling pathways thereby modifying metabolic programs of activated T cells and resulting in altered immune functions. Understanding and therapeutic targeting of metabolic programs impacted by coinhibitory receptors might have significant clinical implications for the treatment of chronic infections, cancer, and autoimmune diseases.
Collapse
Affiliation(s)
- Nikolaos Patsoukis
- Division of Hematology-Oncology, Harvard Medical School, Boston, MA, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jessica D Weaver
- Division of Hematology-Oncology, Harvard Medical School, Boston, MA, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Laura Strauss
- Division of Hematology-Oncology, Harvard Medical School, Boston, MA, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Christoph Herbel
- Division of Hematology-Oncology, Harvard Medical School, Boston, MA, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Pankaj Seth
- Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Vassiliki A Boussiotis
- Division of Hematology-Oncology, Harvard Medical School, Boston, MA, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
96
|
Zhu YP, Yue F, He Y, Li P, Yang Y, Han YT, Zhang YF, Sun GP, Guo DG, Yin M, Wang XN. Prokaryotic expression of the extracellular domain of porcine programmed death 1 (PD-1) and its ligand PD-L1 and identification of the binding with peripheral blood mononuclear cells in vitro. CANADIAN JOURNAL OF VETERINARY RESEARCH = REVUE CANADIENNE DE RECHERCHE VETERINAIRE 2017; 81:147-154. [PMID: 28408783 PMCID: PMC5370541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 12/12/2016] [Indexed: 06/07/2023]
Abstract
Programmed cell death protein 1 (PD-1), a costimulatory molecule of the CD28 family, has 2 ligands, PD-L1 and PD-L2. Our previous studies showed that the expression of PD-1 and PD-L1 is up-regulated during viral infection in pigs. Extensive studies have shown that blockade of the PD-1/PD-L1 pathways by anti-PD-L1 antibody or soluble PD-1 restores exhausted T-cells in humans and mice. In the present study the extracellular domains of PD-1 and PD-L1 were used to evaluate the binding of PD-1 and PD-L1 with peripheral blood mononuclear cells (PBMCs). We amplified the cDNA encoding the extracellular domains of PD-1 and PD-L1 to construct recombinant expression plasmids and obtain soluble recombinant proteins, which were then labeled with fluorescein isothiocyanate (FITC). The His-ExPD-1 and His-ExPD-L1 recombinant proteins were expressed in the form of inclusion bodies with a relative molecular weight of 33.0 and 45.0 kDa, respectively. We then prepared polyclonal antibodies against the proteins with a multi-antiserum titer of 1:102 400. Binding of the proteins with PBMCs was evaluated by flow cytometry. The fluorescence signals of His-ExPD-1-FITC and His-ExPD-L1-FITC were greater than those for the FITC control. These results suggest that the soluble recombinant proteins may be used to prepare monoclonal antibodies to block the PD-1/PD-L1 pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Xuan-Nian Wang
- Address all correspondence to Dr. Xuan-Nian Wang; telephone: +86 373 3682111; fax: +86 373 3683344; e-mail:
| |
Collapse
|
97
|
Kouidhi S, Elgaaied AB, Chouaib S. Impact of Metabolism on T-Cell Differentiation and Function and Cross Talk with Tumor Microenvironment. Front Immunol 2017; 8:270. [PMID: 28348562 PMCID: PMC5346542 DOI: 10.3389/fimmu.2017.00270] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 02/24/2017] [Indexed: 12/12/2022] Open
Abstract
The immune system and metabolism are highly integrated and multilevel interactions between metabolic system and T lymphocyte signaling and fate exist. Accumulating evidence indicates that the regulation of nutrient uptake and utilization in T cells is critically important for the control of their differentiation and manipulating metabolic pathways in these cells can shape their function and survival. This review will discuss some potential cell metabolism pathways involved in shaping T lymphocyte function and differentiation. It will also describe show subsets of T cells have specific metabolic requirements and signaling pathways that contribute to their respective function. Examples showing the apparent similarity between cancer cell metabolism and T cells during activation are illustrated and finally some mechanisms being used by tumor microenvironment to orchestrate T-cell metabolic dysregulation and the subsequent emergence of immune suppression are discussed. We believe that targeting T-cell metabolism may provide an additional opportunity to manipulate T-cell function in the development of novel therapeutics.
Collapse
Affiliation(s)
- Soumaya Kouidhi
- ISBST, Laboratory BVBGR, LR11ES31, Higher Institute of Biotechnology of Sidi Thabet, University of Manouba, Sidi Thabet, Tunisia; Laboratory of Genetics, Immunology and Human Pathology, Faculty of Sciences of Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Amel Benammar Elgaaied
- Laboratory of Genetics, Immunology and Human Pathology, Faculty of Sciences of Tunis, University Tunis El Manar , Tunis , Tunisia
| | - Salem Chouaib
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1186, Laboratory «Integrative Tumor Immunology and Genetic Oncology», Equipe Labellisée LIGUE 2015, Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Gustave Roussy, University of Paris-Sud, Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Gustave Roussy, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
98
|
Efficacy and tolerability of nivolumab after allogeneic transplantation for relapsed Hodgkin lymphoma. Blood 2017; 129:2471-2478. [PMID: 28270452 DOI: 10.1182/blood-2016-11-749556] [Citation(s) in RCA: 177] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 02/24/2017] [Indexed: 11/20/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is indicated for patients with relapsed or refractory Hodgkin lymphoma (HL). Although long-term disease control can be achieved, relapse is still frequent. The programmed cell death protein 1 (PD-1) pathway-blocking antibody nivolumab has shown substantial therapeutic activity and an acceptable safety profile in patients with relapsed or refractory HL who did not receive allo-HCT. However, PD-1 blocking strategy can increase the risk of graft-versus-host disease (GVHD) in murine models. We retrospectively assessed the efficacy and toxicity of nivolumab as a single agent in 20 HL patients relapsing after allo-HCT. GVHD occurred in 6 patients (30%) after nivolumab initiation. All 6 patients had prior history of acute GVHD. The patients with nivolumab-induced GVHD were managed by standard treatment for acute GVHD. Two patients died as a result of GVHD, 1 of progressive disease and 1 of complications related to a second allo-HCT. Overall response rate was 95%. At a median follow-up of 370 days, the 1-year progression-free survival rate was 58.2% (95% CI, 33.1%-76.7%) and the overall survival rate was 78.7% (95% CI, 52.4%-91.5%). Among 13 patients still in response, 6 received a single dose of nivolumab and 7 remain on nivolumab. Compared with standard options for this indication, our results show that nivolumab is effective with an acceptable safety profile.
Collapse
|
99
|
Shad AT, Huo JS, Darcy C, Abu-Ghosh A, Esposito G, Holuba MJ, Robey N, Cooke KR, Symons HJ, Chen AR, Llosa NJ. Tolerance and effectiveness of nivolumab after pediatric T-cell replete, haploidentical, bone marrow transplantation: A case report. Pediatr Blood Cancer 2017; 64. [PMID: 27650634 DOI: 10.1002/pbc.26257] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 08/06/2016] [Accepted: 08/13/2016] [Indexed: 01/28/2023]
Abstract
To date, there has been a lack of pediatric experience regarding the efficacy and tolerability of immune checkpoint inhibitors after haploidentical hematopoietic stem cell transplant (HSCT). We present the case of a 22-year-old female with multiple-relapsed Hodgkin lymphoma (HL) who presented with a new relapse after haploidentical (post-haplo) HSCT. Anti-PD-1 therapy with nivolumab resulted in significant objective disease response and clinical improvement without notable side effects, including the absence of a graft-versus-host disease (GVHD). This case report suggests that immune checkpoint inhibition may be safely tolerated even in the setting of haploidentical HSCT, without triggering overt GVHD.
Collapse
Affiliation(s)
- Aziza T Shad
- Department of Pediatric Hematology/Oncology, The Herman & Walter Samuelson Children's Hospital at Sinai, Baltimore, Maryland.,Department of Pediatric Oncology, Medstar Georgetown University Hospital, Washington, District of Columbia
| | - Jeffrey S Huo
- Department of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Courtney Darcy
- Department of Pediatric Oncology, Medstar Georgetown University Hospital, Washington, District of Columbia
| | - Amal Abu-Ghosh
- Department of Pediatric Oncology, Medstar Georgetown University Hospital, Washington, District of Columbia
| | - Giuseppe Esposito
- Department of Nuclear Medicine, Medstar Georgetown University Hospital, Washington, District of Columbia
| | - Mary-Jo Holuba
- Department of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nancy Robey
- Department of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kenneth R Cooke
- Department of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Heather J Symons
- Department of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Allen R Chen
- Department of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nicolas J Llosa
- Department of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
100
|
Kean LS, Turka LA, Blazar BR. Advances in targeting co-inhibitory and co-stimulatory pathways in transplantation settings: the Yin to the Yang of cancer immunotherapy. Immunol Rev 2017; 276:192-212. [PMID: 28258702 PMCID: PMC5338458 DOI: 10.1111/imr.12523] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the past decade, the power of harnessing T-cell co-signaling pathways has become increasingly understood to have significant clinical importance. In cancer immunotherapy, the field has concentrated on two related modalities: First, targeting cancer antigens through highly activated chimeric antigen T cells (CAR-Ts) and second, re-animating endogenous quiescent T cells through checkpoint blockade. In each of these strategies, the therapeutic goal is to re-ignite T-cell immunity, in order to eradicate tumors. In transplantation, there is also great interest in targeting T-cell co-signaling, but with the opposite goal: in this field, we seek the Yin to cancer immunotherapy's Yang, and focus on manipulating T-cell co-signaling to induce tolerance rather than activation. In this review, we discuss the major T-cell signaling pathways that are being investigated for tolerance induction, detailing preclinical studies and the path to the clinic for many of these molecules. These include blockade of co-stimulation pathways and agonism of coinhibitory pathways, in order to achieve the delicate state of balance that is transplant tolerance: a state which guarantees lifelong transplant acceptance without ongoing immunosuppression, and with preservation of protective immune responses. In the context of the clinical translation of immune tolerance strategies, we discuss the significant challenge that is embodied by the fact that targeted pathway modulators may have opposing effects on tolerance based on their impact on effector vs regulatory T-cell biology. Achieving this delicate balance holds the key to the major challenge of transplantation: lifelong control of alloreactivity while maintaining an otherwise intact immune system.
Collapse
Affiliation(s)
- Leslie S Kean
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA
- The Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Laurence A Turka
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Immune Tolerance Network, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics and the Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|