101
|
Alloantigen expression on malignant cells and healthy host tissue influences graft-versus-tumor reactions after allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant 2018; 53:807-819. [PMID: 29362503 DOI: 10.1038/s41409-017-0071-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 11/27/2017] [Accepted: 11/28/2017] [Indexed: 11/08/2022]
Abstract
Durable remissions of hematological malignancies regularly observed following allogeneic hematopoietic stem cell transplantation (aHSCT) are due to the conditioning regimen, as well as an immunological phenomenon called graft-versus-leukemia (GVL) or graft-versus-tumor (GVT) effect. The development of GVL is closely linked to graft-versus-host disease (GVHD), the main side effect associated with aHSCT. Both, GVHD and GVL are mediated by donor T cells that are initially activated by antigen-presenting cells that present recipient-derived alloantigens in the context of either matched or mismatched MHC class I molecules. Using murine models of aHSCT we show that ubiquitously expressed minor histocompatibility alloantigens (mHAg) are no relevant target for GVT effects. Interestingly, certain ubiquitously expressed MHC alloantigens augmented GVT effects early after transplantation, while others did not. The magnitude of GVT effects correlated with tumor infiltration by CD8+ cytotoxic T cells and tumor cell apoptosis. Furthermore, the immune response underlying GVHD and GVT was oligoclonal, highlighting that immunodominance is an important factor during alloimmune responses. These results emphasize that alloantigen expression on non-hematopoietic tissues can influence GVT effects in a previously unrecognized fashion. These findings bear significance for harnessing optimal GVL effects in patients receiving aHSCT.
Collapse
|
102
|
Kosugi-Kanaya M, Ueha S, Abe J, Shichino S, Shand FHW, Morikawa T, Kurachi M, Shono Y, Sudo N, Yamashita A, Suenaga F, Yokoyama A, Yong W, Imamura M, Teshima T, Matsushima K. Long-Lasting Graft-Derived Donor T Cells Contribute to the Pathogenesis of Chronic Graft-versus-Host Disease in Mice. Front Immunol 2018; 8:1842. [PMID: 29326717 PMCID: PMC5741650 DOI: 10.3389/fimmu.2017.01842] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 12/05/2017] [Indexed: 11/13/2022] Open
Abstract
Chronic graft-versus-host disease (cGVHD) is a major complication in long-term survivors of allogeneic hematopoietic stem cell transplantation (allo-HSCT). Graft-derived T cells (TG) have been implicated in the induction of cGVHD; however, the extent of their contribution to the pathogenesis of cGVHD remains unclear. Using a mouse model of cGVHD, we demonstrate that TG predominate over hematopoietic stem cell-derived T cells generated de novo (THSC) in cGVHD-affected organs such as the liver and lung even at day 63 after allo-HSCT. Persisting TG, in particular CD8+ TG, not only displayed an exhausted or senescent phenotype but also contained a substantial proportion of cells that had the potential to proliferate and produce inflammatory cytokines. Host antigens indirectly presented by donor HSC-derived hematopoietic cells were involved in the maintenance of TG in the reconstituted host. Selective depletion of TG in the chronic phase of disease resulted in the expansion of THSC and thus neither the survival nor histopathology of cGVHD was ameliorated. On the other hand, THSC depletion caused activation of TG and resulted in a lethal TG-mediated exacerbation of GVHD. The findings presented here clarify the pathological role of long-lasting TG in cGVHD.
Collapse
Affiliation(s)
- Mizuha Kosugi-Kanaya
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,CREST, Japan Science and Technology Agency, Tokyo, Japan.,Department of Hematology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Satoshi Ueha
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,CREST, Japan Science and Technology Agency, Tokyo, Japan
| | - Jun Abe
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,CREST, Japan Science and Technology Agency, Tokyo, Japan
| | - Shigeyuki Shichino
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,CREST, Japan Science and Technology Agency, Tokyo, Japan
| | - Francis H W Shand
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,CREST, Japan Science and Technology Agency, Tokyo, Japan
| | - Teppei Morikawa
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Makoto Kurachi
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| | - Yusuke Shono
- Department of Immunology, Memorial Sloan-Kettering Cancer Center, New York, NY, United States
| | - Naoto Sudo
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,CREST, Japan Science and Technology Agency, Tokyo, Japan
| | - Ai Yamashita
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,CREST, Japan Science and Technology Agency, Tokyo, Japan
| | - Fumiko Suenaga
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,CREST, Japan Science and Technology Agency, Tokyo, Japan
| | - Akihiro Yokoyama
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,CREST, Japan Science and Technology Agency, Tokyo, Japan
| | - Wang Yong
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,CREST, Japan Science and Technology Agency, Tokyo, Japan
| | - Masahiro Imamura
- Department of Hematology, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Takanori Teshima
- Department of Hematology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Kouji Matsushima
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,CREST, Japan Science and Technology Agency, Tokyo, Japan
| |
Collapse
|
103
|
Fuchs E. Haploidentical Hematopoietic Cell Transplantation. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00106-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
104
|
Reddy P, Ferrara JL. Graft-Versus-Host Disease and Graft-Versus-Leukemia Responses. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00108-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
105
|
Schroeder MA, Choi J, Staser K, DiPersio JF. The Role of Janus Kinase Signaling in Graft-Versus-Host Disease and Graft Versus Leukemia. Biol Blood Marrow Transplant 2017; 24:1125-1134. [PMID: 29289756 DOI: 10.1016/j.bbmt.2017.12.797] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 12/20/2017] [Indexed: 12/18/2022]
Abstract
For patients with hematologic malignancies, allogeneic hematopoietic cell transplantation (alloHCT) offers a potential curative treatment option, primarily due to an allogeneic immune response against recipient tumor cells (ie, graft-versus-leukemia [GVL] activity). However, many recipients of alloHCT develop graft-versus-host disease (GVHD), in which allogeneic immune responses lead to the damage of healthy tissue. GVHD is a leading cause of nonrelapse mortality and a key contributor to morbidity among patients undergoing alloHCT. Therefore, improving alloHCT outcomes will require treatment strategies that prevent or mitigate GVHD without disrupting GVL activity. Janus kinases (JAKs) are intracellular signaling molecules that are well positioned to regulate GVHD. A variety of cytokines that signal through the JAK signaling pathways play a role in regulating the development, proliferation, and activation of several immune cell types important for GVHD pathogenesis, including dendritic cells, macrophages, T cells, B cells, and neutrophils. Importantly, despite JAK regulation of GVHD, preclinical evidence suggests that JAK inhibition preserves GVL activity. Here we provide an overview of potential roles for JAK signaling in the pathogenesis of acute and chronic GVHD as well as effects on GVL activity. We also review preclinical and clinical results with JAK inhibitors in acute and chronic GVHD settings, with added focus on those actively being evaluated in patients with acute and chronic GVHD.
Collapse
Affiliation(s)
- Mark A Schroeder
- Washington University School of Medicine, Division of Oncology, Section of Stem Cell Transplantation, St. Louis, Missouri.
| | - Jaebok Choi
- Washington University School of Medicine, Division of Oncology, Section of Stem Cell Transplantation, St. Louis, Missouri
| | - Karl Staser
- Washington University School of Medicine, Division of Oncology, Section of Stem Cell Transplantation, St. Louis, Missouri
| | - John F DiPersio
- Washington University School of Medicine, Division of Oncology, Section of Stem Cell Transplantation, St. Louis, Missouri
| |
Collapse
|
106
|
Schneiderman J. Extracorporeal photopheresis: cellular therapy for the treatment of acute and chronic graft-versus-host disease. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2017; 2017:639-644. [PMID: 29222315 PMCID: PMC6142534 DOI: 10.1182/asheducation-2017.1.639] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is a potentially curative option for many disease states. Despite significant improvements in strategies used to prevent and treat acute and chronic graft-versus-host disease (a/cGVHD), they continue to negatively affect outcomes of HSCT significantly. Standard, first-line treatment consists of corticosteroids; beyond this, there is little consistency in therapeutic regimens. Current options include the addition of various immunosuppressive agents, the use of which puts patients at even higher risks for infection and other morbidities. Extracorporeal photopheresis (ECP) is a widely used cellular therapy currently approved by the US Food and Drug Administration for use in patients with cutaneous T-cell lymphoma; it involves the removal of peripherally circulating white blood cells, addition of a light sensitizer, exposure to UV light, and return of the cells to the patient. This results in a series of events ultimately culminating in transition from an inflammatory state to that of tolerance, without global immunosuppression or known long-term adverse effects. Large-scale, prospective studies of the use of ECP in patients with a/cGVHD are necessary in order to develop the optimal treatment regimens.
Collapse
|
107
|
Ali R, Ramdial J, Algaze S, Beitinjaneh A. The Role of Anti-Thymocyte Globulin or Alemtuzumab-Based Serotherapy in the Prophylaxis and Management of Graft-Versus-Host Disease. Biomedicines 2017; 5:biomedicines5040067. [PMID: 29186076 PMCID: PMC5744091 DOI: 10.3390/biomedicines5040067] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 11/01/2017] [Accepted: 11/20/2017] [Indexed: 12/02/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplant is an established treatment modality for hematologic and non-hematologic diseases. However, it is associated with acute and long-term sequelae which can translate into mortality. Graft-versus-host disease (GVHD) remains a glaring obstacle, especially with the advent of reduced-intensity conditioning. Serotherapy capitalizes on antibodies which target T cells and other immune cells to mitigate this effect. This article focuses on the utility of two such agents: anti-thymocyte globulin (ATG) and alemtuzumab. ATG has demonstrated benefit in prophylaxis against GVHD, especially in the chronic presentation. However, there is limited impact of ATG on overall survival and it has little utility in the treatment context. There may be an initial improvement, particularly in skin manifestations, but no substantial benefit has been elicited. Alemtuzumab has shown benefit in both prophylaxis and treatment of GVHD, but at the consequence of a more profound immunosuppressive phase, mandating aggressive viral prophylaxis. There remains heterogeneity in the doses and regimens of the agents, with no standardized protocol in place. Furthermore, it seems that once steroid-refractory GVHD has been established, there is little that can be offered to offset the ultimately dismal outcome. Here we present a systematic overview of ATG- or alemtuzumab-based serotherapy in the prophylaxis and management of GVHD.
Collapse
Affiliation(s)
- Robert Ali
- Hematology/Medical Oncology Fellow, University of Miami/Miller School of Medicine, Miami, FL 33136, USA.
| | - Jeremy Ramdial
- Hematology/Medical Oncology Fellow, University of Miami/Miller School of Medicine, Miami, FL 33136, USA.
| | - Sandra Algaze
- Internal Medicine Residency Program, University of Miami/Miller School of Medicine, Miami, FL 33136, USA.
| | - Amer Beitinjaneh
- Associate Professor of Medicine, Stem Cell Transplant and Cellular Therapy Program, Sylvester Comprehensive Cancer Center, University of Miami/Miller School of Medicine; Miami, FL 33136, USA.
| |
Collapse
|
108
|
Zogas N, Karponi G, Iordanidis F, Malasidis S, Paraskevas V, Papadopoulou A, Scouras ZG, Anagnostopoulos A, Yannaki E. The ex vivo toll-like receptor 7 tolerance induction in donor lymphocytes prevents murine acute graft-versus-host disease. Cytotherapy 2017; 20:149-164. [PMID: 29150086 DOI: 10.1016/j.jcyt.2017.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 08/27/2017] [Accepted: 09/11/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND AIMS Acute graft-versus-host disease (aGVHD) remains a major cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation, mediated by alloreactive donor T cells. Toll-like receptors (TLRs), a family of conserved pattern-recognition receptors (PRRs), represent key players in donors' T-cell activation during aGVHD; however, a regulatory, tolerogenic role for certain TLRs has been recognized in a different context. We investigated whether the ex vivo-induced TLR-2,-4,-7 tolerance in donor cells could prevent alloreactivity in a mismatched transplantation model. METHODS TLR-2,-4,-7 tolerance was induced in mouse splenocytes, after stimulation with low doses of corresponding ligands. Cellular and molecular changes of the TLR-tolerant splenocytes and purified T cells were assessed by immunophenotypic and gene expression analyses. Incidence of aGVHD was evaluated by the clinical score and survival as well as histopathology of target tissues. RESULTS Only the R848-induced TLR7 tolerance prevented aGVHD. The TLR7 ligand-induced tolerance lasted for a critical post-transplant period and was associated with distinct cellular and molecular signatures characterized by induction of regulatory T cells, reduced alloreactivity and balanced regulation of inflammatory signaling and innate immune responses. The TLR7-tolerant T cells preserved the immunological memory and generated in vitro virus-specific T cells upon antigen stimulation. The anti-aGVHD tolerization effect was direct and specific to TLR7 and required the receptor-ligand interaction; TLR7-/- T cells isolated from B6 TLR7-/- mice presented a distinct gene expression profile but failed to prevent aGVHD. DISCUSSION We propose an effective and clinically applicable ex vivo approach for aGVHD prevention through a transient and reversible immune reprogramming exerted by TLR7-tolerant donor lymphocytes.
Collapse
Affiliation(s)
- Nikolaos Zogas
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece; Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Garyfalia Karponi
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece
| | - Fotios Iordanidis
- Cellular Pathology Laboratory, Cheltenham General Hospital, Cheltenham, United Kingdom
| | - Stylianos Malasidis
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece
| | - Vasilios Paraskevas
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece; Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Anastasia Papadopoulou
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece
| | - Zaharias George Scouras
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Achilles Anagnostopoulos
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece
| | - Evangelia Yannaki
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece; Department of Medicine, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
109
|
León-Rodríguez E, Rivera-Franco MM, Gómez-Martín D, Romo-Tena J, Juárez-Vega G, Merayo-Chalico J, Alcocer-Varela J. Differential T cell subsets and cytokine profile between steady-state and G-CSF-primed bone marrow and its association with graft-versus-host disease. Leuk Res 2017; 63:47-52. [PMID: 29101827 DOI: 10.1016/j.leukres.2017.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/19/2017] [Accepted: 10/24/2017] [Indexed: 11/19/2022]
Abstract
Graft-versus-host disease (GVHD) is a major complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT). At our Institution, patients transplanted using G-CSF-primed bone marrow (G-BM), have a lower incidence of GVHD when compared to other sources. The objective of this study was to analyze and compare T cell subsets and cytokines in donor G-BM and steady-state BM (SS-BM). A prospective study was performed in 48 donor samples. Mononuclear cells were isolated by gradient density. T cell subsets and cytokine production in supernatants were analyzed by multiparametric flow cytometry. Six and 16 patients developed acute and chronic GVHD, respectively. Patients who developed GVHD were characterized by a predominant pro-inflammatory response (IL-17A (10.02 vs 0.43pg/mL, p=0.006), TNF-α (54.57 vs 0.81pg/mL, p=0.001)), in contrast to a deficient suppressor profile (IL-10 (7.87 vs 41.37pg/mL, p=0.003)) and Tregs (0.95% vs 1.52%, p=0.004). G-BM showed an enhanced suppressive phenotype (increased Th2 and Tregs) in comparison to SS-BM. GVHD is associated with an imbalance between pro-inflammatory and suppressor immune responses. G-BM showed a more favorable immunologic profile characterized by diminished pro-inflammatory cytokine production, which was associated with a lower frequency of GVHD in our cohort.
Collapse
Affiliation(s)
- Eucario León-Rodríguez
- Stem Cell Transplantation Program, Hematology and Oncology Department, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Vasco de Quiroga 15, Belisario Dominguez Seccion XVI, Tlalpan, 14080, Mexico City, Mexico.
| | - Monica M Rivera-Franco
- Stem Cell Transplantation Program, Hematology and Oncology Department, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico
| | - Diana Gómez-Martín
- Immunology and Rheumatology Department, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico
| | - Jorge Romo-Tena
- Immunology and Rheumatology Department, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico
| | - Guillermo Juárez-Vega
- Flow Cytometry Unit. Red de Apoyo a la Investigación CIC-UNAM, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico
| | - Javier Merayo-Chalico
- Immunology and Rheumatology Department, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico
| | - Jorge Alcocer-Varela
- Immunology and Rheumatology Department, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico
| |
Collapse
|
110
|
Perkey E, Maillard I. New Insights into Graft-Versus-Host Disease and Graft Rejection. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 13:219-245. [PMID: 29099650 DOI: 10.1146/annurev-pathol-020117-043720] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Allogeneic transplantation of foreign organs or tissues has lifesaving potential, but can lead to serious complications. After solid organ transplantation, immune-mediated rejection mandates the use of prolonged global immunosuppression and limits the life span of transplanted allografts. After bone marrow transplantation, donor-derived immune cells can trigger life-threatening graft-versus-host disease. T cells are central mediators of alloimmune complications and the target of most existing therapeutic interventions. We review recent progress in identifying multiple cell types in addition to T cells and new molecular pathways that regulate pathogenic alloreactivity. Key discoveries include the cellular subsets that function as potential sources of alloantigens, the cross talk of innate lymphoid cells with damaged epithelia and with the recipient microbiome, the impact of the alarmin interleukin-33 on alloreactivity, and the role of Notch ligands expressed by fibroblastic stromal cells in alloimmunity. While refining our understanding of transplantation immunobiology, these findings identify new therapeutic targets and new areas of investigation.
Collapse
Affiliation(s)
- Eric Perkey
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, USA;
| | - Ivan Maillard
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, USA; .,Department of Internal Medicine, Division of Hematology-Oncology, and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109, USA.,Department of Medicine, Division of Hematology-Oncology, and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| |
Collapse
|
111
|
Jia H, Cui J, Jia X, Zhao J, Feng Y, Zhao P, Zang D, Yu J, Zhao T, Wang H, Xu K. Therapeutic effects of STAT3 inhibition by nifuroxazide on murine acute graft graft-vs.-host disease: Old drug, new use. Mol Med Rep 2017; 16:9480-9486. [PMID: 29152660 PMCID: PMC5780006 DOI: 10.3892/mmr.2017.7825] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 05/26/2017] [Indexed: 12/30/2022] Open
Abstract
Graft-vs.-host disease (GvHD) is a major and lethal complication of allogeneic bone marrow transplantation (allo-BMT). Although great development has been made, the treatment progress of this disorder is slow. Research has illustrated that STAT3 was critical for T cell alloactivation in GvHD. In the present study, the authors hypothesized that nifuroxazide, as the STAT3 inhibitor, treatment may attenuate the development of acute GvHD (aGvHD). The results demonstrated that nifuroxazide suppressed the development of aGvHD and significantly delayed aGvHD-induced lethality. Mice receiving nifuroxazide had mostly normal-appearing skin with minimal focal ulceration, mild edema and congestion in the liver, and a less-pronounced villus injury and less inflammatory infiltrate in the small intestine. Treatment with nifuroxazide inhibited the activation of STAT3, resulting in the regulation of the CD4+ T cells and CD4+CD25+ T cells and reduction of interferon-γ and tumor necrosis factor-α levels. In conclusion, nifuroxazide may be efficacious for post-transplant of GvHD, providing a potent drug for use as a prophylactic or as a second-line therapy for aGvHD in clinical trials.
Collapse
Affiliation(s)
- Huijie Jia
- Department of Pathology, Xinxiang Medical University, Xinxiang, Henan 453000, P.R. China
| | - Jing Cui
- Department of Pathology, Xinxiang Medical University, Xinxiang, Henan 453000, P.R. China
| | - Xiaolong Jia
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan 453000, P.R. China
| | - Jingjing Zhao
- Department of Immunology, Xinxiang Medical University, Xinxiang, Henan 453000, P.R. China
| | - Yuchen Feng
- Department of Immunology, Xinxiang Medical University, Xinxiang, Henan 453000, P.R. China
| | - Peijuan Zhao
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan 453000, P.R. China
| | - Dan Zang
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453000, P.R. China
| | - Jian Yu
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453000, P.R. China
| | - Tiesuo Zhao
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan 453000, P.R. China
| | - Hui Wang
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan 453000, P.R. China
| | - Kailin Xu
- Laboratory of Transplantation and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
112
|
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
|
113
|
IAPs protect host target tissues from graft-versus-host disease in mice. Blood Adv 2017; 1:1517-1532. [PMID: 29296793 DOI: 10.1182/bloodadvances.2017004242] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 07/01/2017] [Indexed: 12/13/2022] Open
Abstract
Inhibitors of apoptosis proteins (IAPs) regulate apoptosis, but little is known about the role of IAPs in the regulation of immunity. Development of IAP inhibition by second mitochondria-derived activator of caspase (SMAC) mimetics is emerging as a novel therapeutic strategy to treat malignancies. We explored the role of IAPs in allogeneic immunity with 2 distinct yet complementary strategies, namely, chemical and genetic approaches, in clinically relevant models of experimental bone marrow transplantation (BMT). The small-molecule pan-IAP inhibitor SMAC mimetic AT-406 aggravated gastrointestinal graft-versus-host disease (GVHD) in multiple models. The role of specific IAPs in various host and donor cellular compartments was explored by utilizing X-linked IAP (XIAP)- and cellular IAP (cIAP)-deficient animals as donors or recipients. Donor T cells from C57BL/6 cIAP1-/- or XIAP-/- animals demonstrated equivalent GVHD severity and allogeneic responses, both in vivo and in vitro, when compared with B6 wild-type (B6-WT) T cells. By contrast, when used as recipient animals, both XIAP-/- and cIAP1-/- animals demonstrated increased mortality from GVHD when compared with B6-WT animals. BM chimera studies revealed that cIAP and XIAP deficiency in host nonhematopoietic target cells, but not in host hematopoietic-derived cells, is critical for exacerbation of GVHD. Intestinal epithelial cells from IAP-deficient animals showed reduced levels of antiapoptotic proteins as well as autophagy-related protein LC3 after allogeneic BMT. Collectively, our data highlight a novel immune cell-independent but target tissue-intrinsic role for IAPs in the regulation of gastrointestinal damage from GVHD.
Collapse
|
114
|
Toubai T, Rossi C, Oravecz-Wilson K, Zajac C, Liu C, Braun T, Fujiwara H, Wu J, Sun Y, Brabbs S, Tamaki H, Magenau J, Zheng P, Liu Y, Reddy P. Siglec-G represses DAMP-mediated effects on T cells. JCI Insight 2017; 2:92293. [PMID: 28724800 DOI: 10.1172/jci.insight.92293] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 06/08/2017] [Indexed: 12/12/2022] Open
Abstract
The role of negative regulators or suppressors of the damage-associated molecular pattern-mediated (DAMP-mediated) stimulation of innate immune responses is being increasingly appreciated. However, the presence and function of suppressors of DAMP-mediated effects on T cells, and whether they can be targeted to mitigate T cell-dependent immunopathology remain unknown. Sialic acid-binding immunoglobulin-like lectin G (Siglec-G) is a negative regulator of DAMP-mediated responses in innate immune cells, but its T cell-autonomous role is unknown. Utilizing loss-of-function-based (genetic knockout) and gain-of-function-based (agonist) approaches, we demonstrate that in the presence of certain DAMPs, Siglec-G suppressed in vitro and in vivo T cell responses. We also demonstrate that its T cell-autonomous role is critical for modulating the severity of the T cell-mediated immunopathology, graft-versus-host disease (GVHD). Enhancing the Siglec-G signaling in donor T cells with its agonist, a CD24Fc fusion protein, ameliorated GVHD while preserving sufficient graft-versus-tumor (GVT) effects in vivo. Collectively, these data demonstrate that Siglec-G is a potentially novel negative regulator of T cell responses, which can be targeted to mitigate GVHD.
Collapse
Affiliation(s)
- Tomomi Toubai
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Corinne Rossi
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Katherine Oravecz-Wilson
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Cynthia Zajac
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Chen Liu
- Department of Pathology and Laboratory Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Thomas Braun
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, USA
| | - Hideaki Fujiwara
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Julia Wu
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Yaping Sun
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Stuart Brabbs
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Hiroya Tamaki
- Division of Hematology, Department of Internal Medicine, Hyogo College of Medicine, Hyogo, Japan
| | - John Magenau
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Pang Zheng
- Center for Cancer and Immunology Research, Children's National Medical Center, Washington DC, USA
| | - Yang Liu
- Center for Cancer and Immunology Research, Children's National Medical Center, Washington DC, USA
| | - Pavan Reddy
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| |
Collapse
|
115
|
STAT3 Expression in Host Myeloid Cells Controls Graft-versus-Host Disease Severity. Biol Blood Marrow Transplant 2017; 23:1622-1630. [PMID: 28694183 DOI: 10.1016/j.bbmt.2017.06.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 06/27/2017] [Indexed: 02/04/2023]
Abstract
Professional antigen-presenting cells (APCs) are important modulators of acute graft-versus-host disease (GVHD). Although dendritic cells (DCs) are the most potent APC subset, other myeloid cells, especially macrophages (MFs) and neutrophils, recently have been shown to play a role in the severity of GVHD. The critical molecular mechanisms that determine the functions of myeloid cells in GVHD are unclear, however. Signal transducer and activator of transcription 3 (STAT3) is a master transcription factor that plays a crucial role in regulating immunity, but its role in MF biology and in acute GVHD remains unknown. To determine the impact of myeloid cell-specific expression of STAT3 on the severity of acute GVHD, we used myeloid cell-specific STAT3-deficient LysM-Cre/STAT3fl/- animals as recipients and donors in well-characterized experimental models of acute GVHD. We found that reduced expression of STAT3 in myeloid cells from the hosts, but not the donors, increased inflammation, increased donor T cell activation, and exacerbated GVHD. Our data demonstrate that STAT3 in host myeloid cells, such as MFs, dampens acute GVHD.
Collapse
|
116
|
Tormo A, Khodayarian F, Cui Y, Al-Chami E, Kanjarawi R, Noé B, Wang H, Rafei M. Interleukin-21 promotes thymopoiesis recovery following hematopoietic stem cell transplantation. J Hematol Oncol 2017; 10:120. [PMID: 28615039 PMCID: PMC5471903 DOI: 10.1186/s13045-017-0490-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 06/06/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Impaired T cell reconstitution remains a major deterrent in the field of bone marrow (BM) transplantation (BMT) due to pre-conditioning-induced damages inflicted to the thymi of recipient hosts. Given the previously reported thymo-stimulatory property of interleukin (IL)-21, we reasoned that its use post-BMT could have a profound effect on de novo T cell development. METHODS To evaluate the effect of IL-21 on de novo T cell development in vivo, BM derived from RAG2p-GFP mice was transplanted into LP/J mice. Lymphocyte reconstitution was first assessed using a hematological analyzer and a flow cytometer on collected blood samples. Detailed flow cytometry analysis was then performed on the BM, thymus, and spleen of transplanted animals. Finally, the effect of human IL-21 on thymopoiesis was validated in humanized mice. RESULTS Using a major histocompatibility complex (MHC)-matched allogeneic BMT model, we found that IL-21 administration improves immune reconstitution by triggering the proliferation of BM Lin-Sca1+c-kit+ (LSK) subsets. The pharmacological effect of IL-21 also culminates in the recovery of both hematopoietic (thymocytes) and non-hematopoietic (stromal) cells within the thymi of IL-21-treated recipient animals. Although T cells derived from all transplanted groups proliferate, secrete various cytokines, and express granzyme B similarly in response to T cell receptor (TCR) stimulation, full regeneration of peripheral naïve CD4+ and CD8+ T cells and normal TCRvβ distribution could only be detected in IL-21-treated recipient mice. Astonishingly, none of the recipient mice who underwent IL-21 treatment developed graft-versus-host disease (GVHD) in the MHC-matched allogeneic setting while the graft-versus-tumor (GVT) effect was strongly retained. Inhibition of GVHD onset could also be attributed to the enhanced generation of regulatory B cells (B10) observed in the IL-21, but not PBS, recipient mice. We also tested the thymopoiesis-stimulating property of human IL-21 in NSG mice transplanted with cord blood (CB) and found significant improvement in de novo human CD3+ T cell development. CONCLUSIONS In sum, our study indicates that IL-21 represents a new class of unforeseen thymopoietin capable of restoring thymic function following BMT.
Collapse
Affiliation(s)
- Aurélie Tormo
- The Department of Pharmacology and Physiology, Université de Montréal, 2900 Edouard-Montpetit BLVD, Montréal, Québec, H3T 1J4, Canada
| | - Fatemeh Khodayarian
- The Department of Pharmacology and Physiology, Université de Montréal, 2900 Edouard-Montpetit BLVD, Montréal, Québec, H3T 1J4, Canada
| | - Yun Cui
- The Department of Pharmacology and Physiology, Université de Montréal, 2900 Edouard-Montpetit BLVD, Montréal, Québec, H3T 1J4, Canada
| | - Edouard Al-Chami
- The Department of Pharmacology and Physiology, Université de Montréal, 2900 Edouard-Montpetit BLVD, Montréal, Québec, H3T 1J4, Canada
| | - Reem Kanjarawi
- The Department of Pharmacology and Physiology, Université de Montréal, 2900 Edouard-Montpetit BLVD, Montréal, Québec, H3T 1J4, Canada
| | - Beatriz Noé
- The Department of Pharmacology and Physiology, Université de Montréal, 2900 Edouard-Montpetit BLVD, Montréal, Québec, H3T 1J4, Canada
| | - Huijie Wang
- The Department of Pharmacology and Physiology, Université de Montréal, 2900 Edouard-Montpetit BLVD, Montréal, Québec, H3T 1J4, Canada
| | - Moutih Rafei
- The Department of Pharmacology and Physiology, Université de Montréal, 2900 Edouard-Montpetit BLVD, Montréal, Québec, H3T 1J4, Canada. .,The Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, 2900 Edouard-Montpetit BLVD, Montréal, Québec, H3T 1J4, Canada. .,The Department of Microbiology and Immunology, McGill University, 3775 University Street, Montréal, Québec, H3A 2B4, Canada.
| |
Collapse
|
117
|
Franssen LE, Roeven MWH, Hobo W, Doorn R, Oostvogels R, Falkenburg JHF, van de Donk NW, Kester MGD, Fredrix H, Westinga K, Slaper-Cortenbach I, Spierings E, Kersten MJ, Dolstra H, Mutis T, Schaap N, Lokhorst HM. A phase I/II minor histocompatibility antigen-loaded dendritic cell vaccination trial to safely improve the efficacy of donor lymphocyte infusions in myeloma. Bone Marrow Transplant 2017; 52:1378-1383. [PMID: 28581468 DOI: 10.1038/bmt.2017.118] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 04/15/2017] [Accepted: 04/20/2017] [Indexed: 12/22/2022]
Abstract
Allogeneic stem cell transplantation (allo-SCT) with or without donor lymphocyte infusions (DLI) is the only curative option for several hematological malignancies. Unfortunately, allo-SCT is often associated with GvHD, and patients often relapse. We therefore aim to improve the graft-versus-tumor effect, without increasing the risk of GvHD, by targeting hematopoietic lineage-restricted and tumor-associated minor histocompatibility antigens using peptide-loaded dendritic cell (DC) vaccinations. In the present multicenter study, we report the feasibility, safety and efficacy of this concept. We treated nine multiple myeloma patients with persistent or relapsed disease after allo-SCT and a previous DLI, with donor monocyte-derived mHag-peptide-loaded DC vaccinations combined with a second DLI. Vaccinations were well tolerated and no occurrence of GvHD was observed. In five out of nine patients, we were able to show the induction of mHag-specific CD8+ T cells in peripheral blood. Five out of nine patients, of which four developed mHag-specific T cells, showed stable disease (SD) for 3.5-10 months. This study shows that mHag-based donor monocyte-derived DC vaccination combined with DLI is safe, feasible and capable of inducing objective mHag-specific T-cell responses. Future research should focus on further improvement of the vaccination strategy, toward translating the observed T-cell responses into robust clinical responses.
Collapse
Affiliation(s)
- L E Franssen
- Department of Hematology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - M W H Roeven
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - W Hobo
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - R Doorn
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - R Oostvogels
- Department of Hematology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - J H F Falkenburg
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - N W van de Donk
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - M G D Kester
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - H Fredrix
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - K Westinga
- Cell Therapy Facility, University Medical Center Utrecht, Utrecht, The Netherlands
| | - I Slaper-Cortenbach
- Cell Therapy Facility, University Medical Center Utrecht, Utrecht, The Netherlands
| | - E Spierings
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - M J Kersten
- Department of Hematology, Amsterdam Medical Center, Amsterdam, The Netherlands
| | - H Dolstra
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - T Mutis
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - N Schaap
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - H M Lokhorst
- Department of Hematology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
118
|
Ferrara JL, Smith CM, Sheets J, Reddy P, Serody JS. Altered homeostatic regulation of innate and adaptive immunity in lower gastrointestinal tract GVHD pathogenesis. J Clin Invest 2017; 127:2441-2451. [PMID: 28581444 DOI: 10.1172/jci90592] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Lower gastrointestinal (GI) tract graft-versus-host disease (GVHD) is the predominant cause of morbidity and mortality from GVHD after allogeneic stem cell transplantation. Recent data indicate that lower GI tract GVHD is a complicated process mediated by donor/host antigenic disparities. This process is exacerbated by significant changes to the microbiome, and innate and adaptive immune responses that are critical to the induction of disease, persistence of inflammation, and a lack of response to therapy. Here, we discuss new insights into the biology of lower GI tract GVHD and focus on intrinsic pathways and regulatory mechanisms crucial to normal intestinal function. We then describe multiple instances in which these homeostatic mechanisms are altered by donor T cells or conditioning therapy, resulting in exacerbation of GVHD. We also discuss data suggesting that some of these mechanisms produce biomarkers that could be informative as to the severity of GVHD and its response to therapy. Finally, novel therapies that might restore homeostasis in the GI tract during GVHD are highlighted.
Collapse
Affiliation(s)
- James Lm Ferrara
- Departments of Medicine, Pediatrics, and Academic Informatics and Technology, Icahn School of Medicine at Mount Sinai and Tisch Cancer Institute, New York, New York, USA
| | - Christopher M Smith
- Departments of Medicine, Pediatrics, and Academic Informatics and Technology, Icahn School of Medicine at Mount Sinai and Tisch Cancer Institute, New York, New York, USA
| | - Julia Sheets
- University of North Carolina Hospital, Chapel Hill, North Carolina, USA
| | - Pavan Reddy
- Department of Medicine and University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Jonathan S Serody
- Department of Medicine and UNC Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
119
|
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
|
120
|
de Koning C, Admiraal R, Nierkens S, Boelens JJ. Immune reconstitution and outcomes after conditioning with anti-thymocyte-globulin in unrelated cord blood transplantation; the good, the bad, and the ugly. Stem Cell Investig 2017; 4:38. [PMID: 28607912 DOI: 10.21037/sci.2017.05.02] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 04/17/2017] [Indexed: 01/07/2023]
Abstract
Unrelated umbilical cord blood transplantation (UCBT) exhibits a low risk of graft-versus-host-disease (GvHD) and has unique potent anti-virus and anti-leukemia effects. Anti-thymocyte globulin (ATG) in the conditioning regimen for UCBT is successful in reducing graft rejection and GvHD. Nevertheless, this beneficial effect of ATG coincides with its detrimental effect on immune reconstitution. The latter directly relates to a high incidence of viral infections and leukemia relapses. ATG has been used in transplant patients for over 30 years. In recent years, the knowledge on the mechanisms of action of ATG and its implementation in the UCBT setting has increased dramatically. Important data became available showing the highly variable pharmacokinetics (PK) of ATG and its consequence on outcome measures. Here, we review the effects of ATG on immune reconstitution and subsequent outcomes after UCBT, and describe the mechanisms causing these effects. We highlight the importance of optimizing ATG exposure before and after UCBT and discuss strategies to maintain the 'good' and overcome the 'bad and ugly' effects of ATG on UCBT outcome.
Collapse
Affiliation(s)
- Coco de Koning
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Rick Admiraal
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands.,Pediatric Blood and Marrow Transplantation Program, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Stefan Nierkens
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jaap Jan Boelens
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands.,Pediatric Blood and Marrow Transplantation Program, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
121
|
Stickel N, Hanke K, Marschner D, Prinz G, Köhler M, Melchinger W, Pfeifer D, Schmitt-Graeff A, Brummer T, Heine A, Brossart P, Wolf D, von Bubnoff N, Finke J, Duyster J, Ferrara J, Salzer U, Zeiser R. MicroRNA-146a reduces MHC-II expression via targeting JAK/STAT signaling in dendritic cells after stem cell transplantation. Leukemia 2017; 31:2732-2741. [PMID: 28484267 PMCID: PMC6231537 DOI: 10.1038/leu.2017.137] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 04/13/2017] [Accepted: 04/24/2017] [Indexed: 02/07/2023]
Abstract
Acute Graft-versus-host disease (GVHD) is a major immunological complication after allogeneic hematopoietic cell transplantation and a better understanding of the molecular regulation of the disease could help to develop novel targeted therapies. Here we found that a G/C polymorphism within the human microRNA-146a (miR-146a) gene of transplant-recipients, which causes reduced miR-146a levels, was strongly associated with the risk of developing severe acute GVHD (n=289). In mice, deficiency of miR-146a in the hematopoietic system or transfer of recipient-type miR 146a-/- dendritic cells (DCs) enhanced GVHD, while miR-146a mimic-transfected-DCs ameliorated disease. Mechanistically, lack of miR-146a enhanced JAK2 STAT1-pathway activity, which led to higher expression of class II-transactivator (CIITA) and consecutively increased MHCII-levels on DCs. Inhibition of JAK1/2 or CIITA knockdown in DCs prevented miR-146a-/- DC-induced GVHD exacerbation. Consistent with our findings in mice, patients with the miR-146a polymorphism rs2910164 in hematopoietic cells displayed higher MHCII levels on monocytes, which could be targeted by JAK1/2-inhibition. Our findings indicate that the miR-146a polymorphism rs2910164 identifies patients at high risk for GVHD before allo HCT. Functionally we show that miR-146a acts as a central regulator of recipient-type DC activation during GVHD by dampening the pro-inflammatory JAK-STAT/CIITA/MHCII axis, which provides a scientific rationale for early JAK1/2-inhibition in selected patients.
Collapse
Affiliation(s)
- N Stickel
- Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine, ALU Freiburg, Germany.,Faculty of Biology, ALU Freiburg, Germany
| | - K Hanke
- Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, ALU Freiburg, Germany
| | - D Marschner
- Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - G Prinz
- Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - M Köhler
- Spemann Graduate School of Biology and Medicine, ALU Freiburg, Germany.,Faculty of Biology, ALU Freiburg, Germany.,Signal Transduction in Tumour Development and Drug Resistance Group, Institute of Molecular Medicine and Cell Research (IMMZ), ALU Freiburg, Germany
| | - W Melchinger
- Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - D Pfeifer
- Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - A Schmitt-Graeff
- Department of Pathology, Freiburg University Medical Center, ALU Freiburg, Germany
| | - T Brummer
- Signal Transduction in Tumour Development and Drug Resistance Group, Institute of Molecular Medicine and Cell Research (IMMZ), ALU Freiburg, Germany.,Centre for Biological Signaling Studies BIOSS, ALU Freiburg, Germany
| | - A Heine
- Medical Clinic III, Oncology, Hematology and Rheumatology, University Hospital Bonn (UKB), Bonn, Germany
| | - P Brossart
- Medical Clinic III, Oncology, Hematology and Rheumatology, University Hospital Bonn (UKB), Bonn, Germany
| | - D Wolf
- Medical Clinic III, Oncology, Hematology and Rheumatology, University Hospital Bonn (UKB), Bonn, Germany
| | - N von Bubnoff
- Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - J Finke
- Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - J Duyster
- Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - J Ferrara
- Department of Medicine, Hematology and Medical Oncology, Mount Sinai Hospital, New York, NY, USA
| | - U Salzer
- Center for Chronic Immunodeficiency, University Medical Center Freiburg and University of Freiburg, Germany.,Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Germany
| | - R Zeiser
- Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Centre for Biological Signaling Studies BIOSS, ALU Freiburg, Germany.,German Cancer Consortium (DKTK), Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
122
|
Kanate AS, Hari PN, Pasquini MC, Visotcky A, Ahn KW, Boyd J, Guru Murthy GS, Rizzo JD, Saber W, Drobyski W, Michaelis L, Atallah E, Carlson KS, D'Souza A, Fenske TS, Cumpston A, Bunner P, Craig M, Horowitz MM, Hamadani M. Recipient Immune Modulation with Atorvastatin for Acute Graft-versus-Host Disease Prophylaxis after Allogeneic Transplantation. Biol Blood Marrow Transplant 2017; 23:1295-1302. [PMID: 28412518 DOI: 10.1016/j.bbmt.2017.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 04/09/2017] [Indexed: 11/16/2022]
Abstract
Atorvastatin administration to both the donors and recipients of matched related donor (MRD) allogeneic hematopoietic cell transplantation (allo-HCT) as acute graft-versus-host disease (GVHD) prophylaxis has been shown to be safe and effective. However, its efficacy as acute GVHD prophylaxis when given only to allo-HCT recipients is unknown. We conducted a phase II study to evaluate the safety and efficacy of atorvastatin-based acute GVHD prophylaxis given only to the recipients of MRD (n = 30) or matched unrelated donor (MUD) (n = 39) allo-HCT, enrolled in 2 separate cohorts. Atorvastatin (40 mg/day) was administered along with standard GVHD prophylaxis consisting of tacrolimus and methotrexate. All patients were evaluable for acute GVHD. The cumulative incidences of grade II to IV acute GVHD at day +100 in the MRD and MUD cohorts were 9.9% (95% confidence interval [CI], 0 to 20%) and 29.6% (95% CI,15.6% to 43.6%), respectively. The cumulative incidences of grade III and IV acute GVHD at day +100 in the MRD and MUD cohorts were 3.4% (95% CI, 0 to 9.7%) and 18.3% (95% CI, 6.3% to 30.4%), respectively. The corresponding rates of moderate/severe chronic GVHD at 1 year were 28.1% (95% CI, 11% to 45.2%) and 38.9% (95% CI, 20.9% to 57%), respectively. In the MRD cohort, the 1-year nonrelapse mortality, relapse rate, progression-free survival, and overall survival were 6.7% (95% CI, 0 to 15.4%), 43.3% (95% CI, 24.9% to 61.7%), 50% (95% CI, 32.1% to 67.9%), and 66.7% (95% CI, 49.8% to 83.6%), respectively. The respective figures for the MUD cohort were 10.3% (95% CI, 8% to 19.7%), 20.5% (95% CI, 7.9% to 33.1%), 69.2% (95% CI, 54.7% to 83.7%), and 79.5% (95% CI, 66.8% to 92.2%), respectively. No grade 4 toxicities attributable to atorvastatin were seen. In conclusion, the addition of atorvastatin to standard GVHD prophylaxis in only the recipients of MRD and MUD allo-HCT appears to be feasible and safe. The preliminary efficacy seen here warrants confirmation in randomized trials.
Collapse
Affiliation(s)
- Abraham S Kanate
- Osborn Hematopoietic Malignancy and Transplantation Program, West Virginia University, Morgantown, West Virginia
| | - Parameswaran N Hari
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Marcelo C Pasquini
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Alexis Visotcky
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Kwang W Ahn
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jennifer Boyd
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | - J Douglas Rizzo
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Wael Saber
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - William Drobyski
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Laura Michaelis
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Ehab Atallah
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Karen S Carlson
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Anita D'Souza
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Timothy S Fenske
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Aaron Cumpston
- Osborn Hematopoietic Malignancy and Transplantation Program, West Virginia University, Morgantown, West Virginia
| | - Pamela Bunner
- Osborn Hematopoietic Malignancy and Transplantation Program, West Virginia University, Morgantown, West Virginia
| | - Michael Craig
- Osborn Hematopoietic Malignancy and Transplantation Program, West Virginia University, Morgantown, West Virginia
| | - Mary M Horowitz
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Mehdi Hamadani
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin.
| |
Collapse
|
123
|
Gaignage M, Marillier RG, Uyttenhove C, Dauguet N, Saxena A, Ryffel B, Michiels T, Coutelier JP, Van Snick J. Mouse nidovirus LDV infection alleviates graft versus host disease and induces type I IFN-dependent inhibition of dendritic cells and allo-responsive T cells. IMMUNITY INFLAMMATION AND DISEASE 2017; 5:200-213. [PMID: 28474504 PMCID: PMC5418140 DOI: 10.1002/iid3.157] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 01/20/2017] [Accepted: 02/08/2017] [Indexed: 11/09/2022]
Abstract
Introduction Viruses have developed multiple mechanisms to alter immune reactions. In 1969, it was reported that lactate dehydrogenase‐elevating virus (LDV), a single stranded positive sense mouse nidovirus, delays skin allograft rejection and inhibits spleen alterations in graft versus host disease (GVHD). As the underlying mechanisms have remained unresolved and given the need for new therapies of this disease, we reassessed the effects of the virus on GVHD and tried to uncover its mode of action. Methods GVHD was induced by transfer of parent (B6) spleen cells to non‐infected or LDV‐infected B6D2F1 recipients. In vitro mixed‐lymhocyte culture (MLC) reactions were used to test the effects of the virus on antigen‐presenting cells (APC) and responder T cells. Results LDV infection resulted in a threefold increase in survival rate with reduced weight loss and liver inflammation but with the establishment of permanent chimerism that correlated with decreased interleukine (IL)‐27 and interferon (IFN)γ plasma levels. Infected mice showed a transient elimination of splenic CD11b+ and CD8α+ conventional dendritic cells (cDCs) required for allogeneic CD4 and CD8 T cell responses in vitro. This drop of APC numbers was not observed with APCs derived from toll‐like receptor (TLR)7‐deficient mice. A second effect of the virus was a decreased T cell proliferation and IFNγ production during MLC without detectable changes in Foxp3+ regulatory T cell (Tregs) numbers. Both cDC and responder T cell inhibition were type I IFN dependent. Although the suppressive effects were very transient, the GVHD inhibition was long‐lasting. Conclusion A type I IFN‐dependent suppression of DC and T cells just after donor spleen cell transplantation induces permanent chimerism and donor cell implantation in a parent to F1 spleen cell transplantation model. If this procedure can be extended to full allogeneic bone marrow transplantation, it could open new therapeutic perspectives for hematopoietic stem cell transplantation (HSCT).
Collapse
Affiliation(s)
- Mélanie Gaignage
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Reece G Marillier
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | | | - Nicolas Dauguet
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Anubha Saxena
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Bernard Ryffel
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), University of Orleans, Orleans, France.,Institute of Infectious Disease and Molecular Medicine, RSA, University of Cape Town, Cape Town, South Africa
| | - Thomas Michiels
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | | | - Jacques Van Snick
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium.,Ludwig Cancer Research, Brussels Branch, Brussels, Belgium
| |
Collapse
|
124
|
Chung J, Ebens CL, Perkey E, Radojcic V, Koch U, Scarpellino L, Tong A, Allen F, Wood S, Feng J, Friedman A, Granadier D, Tran IT, Chai Q, Onder L, Yan M, Reddy P, Blazar BR, Huang AY, Brennan TV, Bishop DK, Ludewig B, Siebel CW, Radtke F, Luther SA, Maillard I. Fibroblastic niches prime T cell alloimmunity through Delta-like Notch ligands. J Clin Invest 2017; 127:1574-1588. [PMID: 28319044 PMCID: PMC5373885 DOI: 10.1172/jci89535] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 01/05/2017] [Indexed: 12/31/2022] Open
Abstract
Alloimmune T cell responses induce graft-versus-host disease (GVHD), a serious complication of allogeneic bone marrow transplantation (allo-BMT). Although Notch signaling mediated by Delta-like 1/4 (DLL1/4) Notch ligands has emerged as a major regulator of GVHD pathogenesis, little is known about the timing of essential Notch signals and the cellular source of Notch ligands after allo-BMT. Here, we have shown that critical DLL1/4-mediated Notch signals are delivered to donor T cells during a short 48-hour window after transplantation in a mouse allo-BMT model. Stromal, but not hematopoietic, cells were the essential source of Notch ligands during in vivo priming of alloreactive T cells. GVHD could be prevented by selective inactivation of Dll1 and Dll4 in subsets of fibroblastic stromal cells that were derived from chemokine Ccl19-expressing host cells, including fibroblastic reticular cells and follicular dendritic cells. However, neither T cell recruitment into secondary lymphoid organs nor initial T cell activation was affected by Dll1/4 loss. Thus, we have uncovered a pathogenic function for fibroblastic stromal cells in alloimmune reactivity that can be dissociated from their homeostatic functions. Our results reveal what we believe to be a previously unrecognized Notch-mediated immunopathogenic role for stromal cell niches in secondary lymphoid organs after allo-BMT and define a framework of early cellular and molecular interactions that regulate T cell alloimmunity.
Collapse
Affiliation(s)
- Jooho Chung
- Graduate Program in Cellular and Molecular Biology
- Life Sciences Institute, and
| | - Christen L. Ebens
- Life Sciences Institute, and
- Division of Hematology-Oncology, Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Eric Perkey
- Graduate Program in Cellular and Molecular Biology
- Life Sciences Institute, and
| | - Vedran Radojcic
- Life Sciences Institute, and
- Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Ute Koch
- École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | | | - Alexander Tong
- Medical Scientist Training Program and Division of Pediatric Hematology-Oncology, Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Frederick Allen
- Medical Scientist Training Program and Division of Pediatric Hematology-Oncology, Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Sherri Wood
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Jiane Feng
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | | - Qian Chai
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Lucas Onder
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Minhong Yan
- Genentech, South San Francisco, California, USA
| | - Pavan Reddy
- Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Bruce R. Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Alex Y. Huang
- Medical Scientist Training Program and Division of Pediatric Hematology-Oncology, Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Todd V. Brennan
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - D. Keith Bishop
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | | | - Freddy Radtke
- École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Sanjiv A. Luther
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Ivan Maillard
- Life Sciences Institute, and
- Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
125
|
Ghimire S, Weber D, Mavin E, Wang XN, Dickinson AM, Holler E. Pathophysiology of GvHD and Other HSCT-Related Major Complications. Front Immunol 2017; 8:79. [PMID: 28373870 PMCID: PMC5357769 DOI: 10.3389/fimmu.2017.00079] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 01/17/2017] [Indexed: 12/13/2022] Open
Abstract
For over 60 years, hematopoietic stem cell transplantation has been the major curative therapy for several hematological and genetic disorders, but its efficacy is limited by the secondary disease called graft versus host disease (GvHD). Huge advances have been made in successful transplantation in order to improve patient quality of life, and yet, complete success is hard to achieve. This review assimilates recent updates on pathophysiology of GvHD, prophylaxis and treatment of GvHD-related complications, and advances in the potential treatment of GvHD.
Collapse
Affiliation(s)
- Sakhila Ghimire
- Department of Internal Medicine III, University Medical Centre , Regensburg , Germany
| | - Daniela Weber
- Department of Internal Medicine III, University Medical Centre , Regensburg , Germany
| | - Emily Mavin
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University , Newcastle , UK
| | - Xiao Nong Wang
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University , Newcastle , UK
| | - Anne Mary Dickinson
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University , Newcastle , UK
| | - Ernst Holler
- Department of Internal Medicine III, University Medical Centre , Regensburg , Germany
| |
Collapse
|
126
|
Gao L, Liu J, Zhang Y, Chen X, Gao L, Zhang C, Liu Y, Kong P, Zhong J, Sun A, Du X, Su Y, Li H, Liu H, Peng X, Zhang X. Low incidence of acute graft-versus-host disease with short-term tacrolimus in haploidentical hematopoietic stem cell transplantation. Leuk Res 2017; 57:27-36. [PMID: 28273549 DOI: 10.1016/j.leukres.2017.02.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 01/22/2017] [Accepted: 02/22/2017] [Indexed: 01/29/2023]
Abstract
Although tacrolimus (Tac) has immunosuppressive properties and exhibits promising efficacy against graft-versus-host disease (GVHD), little is known about Tac in the prophylaxis of GVHD after HLA-haploidentical hematopoietic stem cell transplantation (haplo-HSCT). In a multicenter randomized controlled trial, 174 patients received haplo-HSCT with GVHD prophylaxis involving short-term Tac (from -8days to +30days) or cyclosporine (CsA). The 100day cumulative incidences of acute GVHD (aGVHD) and grade III-IV aGVHD with the short-term Tac regimen and CsA regimen were 29.1 (19.5-38.7)% vs. 50.0(39.6-60.4)% (p=0.005) and 3.6(0.0-7.5)% vs. 13.5(6.1-20.9)% (p=0.027), respectively. There were no significant differences in the incidences of chronic GVHD (cGVHD), relapse and cytomegalovirus infection. Lymphocyte subset analysis showed that T cells decreased to lower levels on the short-term Tac regimen within 3 months of transplantation. The disease-free survival and overall survival on the short-term Tac and CsA regimens were 59.3 (48.9-69.7)% vs. 55.7 (45.3-66.1)% (p=0.696) and 65.1 (55.1-75.1)% vs. 61.4 (51.2-71.6)% (p=0.075), respectively. Our findings indicate that the short-term Tac regimen for GVHD prophylaxis in patients undergoing haplo-HSCT is associated with a low incidence and slight severity of aGVHD and did not increase the incidence of relapse and cytomegalovirus infection.
Collapse
Affiliation(s)
- Lei Gao
- Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
| | - Jia Liu
- Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yanqi Zhang
- Department of Health Statistics, College of Military Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Xinghua Chen
- Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Li Gao
- Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Cheng Zhang
- Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yao Liu
- Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Peiyan Kong
- Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Jiangfan Zhong
- Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, China; Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Aihua Sun
- Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Xin Du
- Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yi Su
- Department of Hematology, General Hospital of Chengdu Military Region of PLA, Chengdu, China
| | - Huimin Li
- Department of Hematology, Affiliated Hospital of Kunming Medical College, Kunming, China
| | - Hong Liu
- Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Xiangui Peng
- Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Xi Zhang
- Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
127
|
Novel Treatment of Chronic Graft-Versus-Host Disease in Mice Using the ER Stress Reducer 4-Phenylbutyric Acid. Sci Rep 2017; 7:41939. [PMID: 28165054 PMCID: PMC5292729 DOI: 10.1038/srep41939] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 01/04/2017] [Indexed: 12/20/2022] Open
Abstract
Chronic graft-versus-host disease (cGVHD) is a notorious complication of allogeneic hematopoietic stem cell transplantation and causes disabling systemic inflammation and fibrosis. In this novel study, we focused on a relationship between endoplasmic reticulum (ER) stress and cGVHD, and aimed to create effective treatment of cGVHD. A series of experiments were conducted using a mouse model of cGVHD. Our data suggested (1) that ER stress was elevated in organs affected by cGVHD and (2) that 4-phenylbutyric acid (PBA) could reduce cGVHD-induced ER stress and thereby alleviate systemic inflammation and fibrosis. Because fibroblasts are thought to be implicated in cGVHD-elicited fibrosis and because macrophages are reported to play a role in the development of cGVHD, we investigated cGVHD-triggered ER stress in fibroblasts and macrophages. Our investigation demonstrated (1) that indicators for ER stress and activation markers for fibroblasts were elevated in cGVHD-affected lacrimal gland fibroblasts and (2) that they could be reduced by PBA. Our work also indicated that splenic macrophages from PBA-dosed mice exhibited the lower levels of ER stress and M2 macrophage markers than those from cGVHD-affected mice. Collectively, this study suggests that the reduction of ER stress utilizing PBA can be a clinically translatable method to treat systemic cGVHD.
Collapse
|
128
|
Zadik Y, Elad S, Shapira A, Shapira MY. Treatment of oral mucosal manifestations of chronic graft-versus-host disease: dexamethasone vs. budesonide. Expert Opin Pharmacother 2017; 18:235-242. [PMID: 28081677 DOI: 10.1080/14656566.2017.1282464] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The oral mucosa is commonly involved in chronic graft-versus-host disease (cGVHD). Oral mucosal cGVHD markedly affect individual's daily function and wellbeing. In some cases, it might become a life threating complication. Areas covered: This article describes the rationale for treatment, method of topical application in the oral cavity, evidence supporting the topical administration of dexamethasone and budesonide for oral cGVHD, and their adverse effects. Expert opinion: Evidence supports the use of topical dexamethasone and budesonide for treatment of oral cGVHD. Topical corticosteroid choice for oral cGVHD, takes into consideration the potency, bioavailability, preferred concentration, and possible adverse effects. Budesonide's pharmacological characteristics mark it as a preferable topical agent for oral cGVHD.
Collapse
Affiliation(s)
- Yehuda Zadik
- a Department of Oral Medicine, Sedation and Maxillofacial Imaging , Hadassah-Hebrew University School of Dental Medicine , Jerusalem , Israel.,b Department of Oral Medicine , The Oral and Maxillofacial Center, Medical Corps, Israel Defense Forces , Tel Hashomer , Israel
| | - Sharon Elad
- c Division of Oral Medicine, Hospital General Dentistry, Eastman Institute for Oral Health , University of Rochester Medical Center , Rochester , NY , USA.,d Wilmot Cancer Center , University of Rochester Medical Center , Rochester , NY , USA
| | - Anat Shapira
- e Pharmacy Division , Hadassah-Hebrew University Medical Center , Jerusalem , Israel
| | - Michael Y Shapira
- f Bone Marrow Transplantation and Cancer Immunotherapy Department , Hadassah-Hebrew University Medical Center , Jerusalem , Israel
| |
Collapse
|
129
|
Ikegame K, Kaida K, Yoshihara S, Yoshihara K, Ishii S, Inoue T, Okada M, Tamaki H, Soma T, Kusunoki Y, Kojima H, Saji H, Ogawa H. Spousal hematopoietic stem cell transplantation. Int J Hematol 2016; 105:646-657. [PMID: 28013483 DOI: 10.1007/s12185-016-2168-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/16/2016] [Accepted: 12/16/2016] [Indexed: 01/21/2023]
Abstract
We report a pilot series of five patients who received stem cell transplantation (SCT) from a spouse for post-transplant relapse or rejection. The inclusion criterion regarding HLA disparities was three or fewer antigen mismatches in the graft-versus-host direction at the HLA-A, B, and DR loci. Four patients received spousal SCT as a third transplant attempt after post-transplant relapse and one as rescue for graft rejection. The reduced intensity conditioning (RIC) regimen consisted of fludarabine, melphalan, and anti-thymocyte globulin (ATG) with 3 Gy of total body irradiation (TBI) for relapse cases and ATG plus 4 Gy of TBI for the rejection case. Graft-versus-host disease (GVHD) prophylaxis consisted of tacrolimus, methylprednisolone, and mycophenolate mofetil. Peripheral blood stem cells were transplanted. Granulocyte engraftment was achieved in all cases between days 9 and 11 (median, 10) with complete spousal chimerism. In three of the five patients, no acute GVHD was observed, while one case developed grade III GVHD and one case grade IV. All four patients evaluable for the anti-leukemic effect achieved complete remission; however, all relapsed between 106 and 334 day post-transplant, and died between days 152 and 548. We suggest that spousal SCT can be performed as a repetitive SCT using a RIC regimen with low-dose ATG and steroid-containing GVHD prophylaxis.
Collapse
Affiliation(s)
- Kazuhiro Ikegame
- Division of Hematology, Department of Internal Medicine, Hyogo Medical College, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan.
| | - Katsuji Kaida
- Division of Hematology, Department of Internal Medicine, Hyogo Medical College, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Satoshi Yoshihara
- Division of Hematology, Department of Internal Medicine, Hyogo Medical College, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan.,Department of Transfusion Medicine and Cell Therapy, Hyogo Medical College, Nishinomiya, Hyogo, Japan
| | - Kyoko Yoshihara
- Division of Hematology, Department of Internal Medicine, Hyogo Medical College, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan.,Department of Transfusion Medicine and Cell Therapy, Hyogo Medical College, Nishinomiya, Hyogo, Japan
| | - Shinichi Ishii
- Division of Hematology, Department of Internal Medicine, Hyogo Medical College, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Takayuki Inoue
- Division of Hematology, Department of Internal Medicine, Hyogo Medical College, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Masaya Okada
- Division of Hematology, Department of Internal Medicine, Hyogo Medical College, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Hiroya Tamaki
- Division of Hematology, Department of Internal Medicine, Hyogo Medical College, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Toshihiro Soma
- Division of Hematology, Department of Internal Medicine, Hyogo Medical College, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | | | | | | | - Hiroyasu Ogawa
- Division of Hematology, Department of Internal Medicine, Hyogo Medical College, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| |
Collapse
|
130
|
Affiliation(s)
- Francine M. Foss
- Bone Marrow Transplantation and Experimental Therapeutics, Tufts New England Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
131
|
Jia H, Zhao T, Ji Y, Jia X, Ren W, Li C, Li M, Xiao Y, Wang H, Xu K. Combined nifuroxazide and SAT05f therapy reduces graft-versus-host disease after experimental allogeneic bone marrow transplantation. Cell Death Dis 2016; 7:e2507. [PMID: 27906171 PMCID: PMC5261008 DOI: 10.1038/cddis.2016.399] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 10/27/2016] [Accepted: 10/31/2016] [Indexed: 01/10/2023]
Abstract
Acute graft-versus-host disease (aGvHD) is the major barrier to the broader use of allogenetic hematopoietic stem cells. However, currently these are no highly specific and efficient drugs. Monotherapy is not sufficient and more efficient and safe therapeutic regimen are urgent need. Studies demonstrated TLR9 and Stat3 signal pathways are critical for antigen-presenting cell maturation and T-cell activation, which are important mediators in aGvHD. Specific block these two critical signal pathways using their inhibitors SAT05f and nifuroxazide may be the novel strategies for aGvHD therapy. The results showed combined therapy significantly decreased the severity of aGvHD and prolonged the survival rate. Furthermore, after treatment, the activation of CD4+ effect T cells was reduced, whereas Treg cells was increased, and the cytokine release was inhibited. In conclusion, combined therapy of nifuroxazide with SAT05f may be potential for the prevention or treatment of aGvHD, providing theoretic and experimental basis.
Collapse
Affiliation(s)
- Huijie Jia
- Department of Pathology, Xinxiang Medical University, Xinxiang 453000, Henan, China
| | - Tiesuo Zhao
- Research Center for Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Yinghua Ji
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan, China
| | - Xiaolong Jia
- Research Center for Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Wenjing Ren
- Department of Dermatology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan, China
| | - Chen Li
- Research Center for Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Minming Li
- Research Center for Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Yali Xiao
- Research Center for Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Hui Wang
- Research Center for Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Kailin Xu
- Laboratory of Transplantation and Immunology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| |
Collapse
|
132
|
Dodge J, Stephans A, Lai J, Drobyski WR, Chen X. Effects of Donor Vitamin A Deficiency and Pharmacologic Modulation of Donor T Cell Retinoic Acid Pathway on the Severity of Experimental Graft-versus-Host Disease. Biol Blood Marrow Transplant 2016; 22:2141-2148. [PMID: 27596131 DOI: 10.1016/j.bbmt.2016.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/01/2016] [Indexed: 02/08/2023]
Abstract
Graft-versus-host disease (GVHD) is the major cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (HSCT). A combination of genetic and nongenetic factors dictates the incidence and severity of GVHD. Recent studies have identified the potential role of the retinoic acid (RA)/retinoic acid receptor (RAR) pathway in the pathogenesis of GVHD. RA is the active metabolite of vitamin A. Thus, a clinically relevant question is whether HSCT donor and/or recipient vitamin A status affects the development of GVHD. It has been previously reported that recipient vitamin A deficiency is associated with reduced intestinal GVHD and prolonged overall survival after experimental allogeneic HSCT. However, it is still unknown whether donor vitamin A status influences GVHD development. In the current study, we report that chronic vitamin A deficiency changes the composition of T cell compartment of donor mice with a reduction in the percentage of CD4+ T cells. We showed that although vitamin A deficiency does not affect donor T cell alloreactivity on a per cell basis, a decreased proportion of donor CD4+ T cells in marrow graft inoculums leads to reduced incidence and severity of GVHD. Furthermore, our proof of principle studies using a pan-RAR antagonist demonstrated that transient inhibition of donor T cell RAR signaling can reduce T cell alloreactivity and their ability to cause lethal GVHD. Our studies provide preclinical evidence that donor vitamin A deficiency may be a nongenetic factor that can modulate the severity of GVHD and pharmacologic interfering RA/RAR pathway in donor T cells might be a valuable approach for mitigating GVHD after allogeneic HSCT.
Collapse
Affiliation(s)
- Joseph Dodge
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Allison Stephans
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jinping Lai
- Department of Pathology, St. Louis University School of Medicine, St. Louis, Missouri
| | - William R Drobyski
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Xiao Chen
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin.
| |
Collapse
|
133
|
Graft-versus-host disease targets ovary and causes female infertility in mice. Blood 2016; 129:1216-1225. [PMID: 27903524 DOI: 10.1182/blood-2016-07-728337] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 11/20/2016] [Indexed: 01/19/2023] Open
Abstract
Infertility associated with ovarian failure is a serious late complication for female survivors of allogeneic hematopoietic stem cell transplantation (SCT). Although pretransplant conditioning regimen has been appreciated as a cause of ovarian failure, increased application of reduced-intensity conditioning allowed us to revisit other factors possibly affecting ovarian function after allogeneic SCT. We have addressed whether donor T-cell-mediated graft-versus-host disease (GVHD) could be causally related to female infertility in mice. Histological evaluation of the ovaries after SCT demonstrated donor T-cell infiltration in close proximity to apoptotic granulosa cells in the ovarian follicles, resulting in impaired follicular hormone production and maturation of ovarian follicles. Mating experiments showed that female recipients of allogeneic SCT deliver significantly fewer newborns than recipients of syngeneic SCT. GVHD-mediated ovary insufficiency and infertility were independent of conditioning. Pharmacologic GVHD prophylaxis protected the ovary from GVHD and preserved fertility. These results demonstrate for the first time that GVHD targets the ovary and impairs ovarian function and fertility and has important clinical implications in young female transplant recipients with nonmalignant diseases, in whom minimally toxic regimens are used.
Collapse
|
134
|
SHP-1 Acts as a Key Regulator of Alloresponses by Modulating LFA-1-Mediated Adhesion in Primary Murine T Cells. Mol Cell Biol 2016; 36:3113-3127. [PMID: 27697866 DOI: 10.1128/mcb.00294-16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/28/2016] [Indexed: 11/20/2022] Open
Abstract
The clinical potential of transplantation is often reduced by T cell-mediated alloresponses that cause graft rejection or graft-versus-host disease. Integrin-mediated adhesion between alloreactive T cells and antigen-presenting cells is essential for allorejection. The identity of the signaling events needed for the activation of integrins such as LFA-1 is poorly understood. Here, we identified a novel role of the protein tyrosine phosphatase SHP-1 in the regulation of murine LFA-1-mediated adhesion in an allograft setting. Upon alloactivation, SHP-1 activity is reduced, resulting in an increase in LFA-1 adhesion compared to that for syngeneically activated T cells. The importance of these differential activation properties was further indicated by small interfering RNA (siRNA) knockdown of SHP-1 in syngeneically and allogeneically stimulated T cells. Mechanistically, SHP-1 modulated the binding of SLP-76 to ADAP by dephosphorylation of the YDGI tyrosine motif of ADAP, a known docking site for the Src family kinase Fyn. This novel key role of SHP-1 in the regulation of LFA-1-mediated adhesion may provide a new insight into T cell-mediated alloresponses and may pave the way to the development of new immunosuppressive pharmaceutical agents.
Collapse
|
135
|
Impola U, Larjo A, Salmenniemi U, Putkonen M, Itälä-Remes M, Partanen J. Graft Immune Cell Composition Associates with Clinical Outcome of Allogeneic Hematopoietic Stem Cell Transplantation in Patients with AML. Front Immunol 2016; 7:523. [PMID: 27917176 PMCID: PMC5117118 DOI: 10.3389/fimmu.2016.00523] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 11/09/2016] [Indexed: 12/17/2022] Open
Abstract
Complications of allogeneic hematopoietic stem cell transplantation (HSCT) have been attributed to immune cells transferred into the patient with the graft. However, a detailed immune cell composition of the graft is usually not evaluated. In the present study, we determined the level of variation in the composition of immune cells between clinical HSCT grafts and whether this variation is associated with clinical outcome. Sizes of major immune cell populations in 50 clinical grafts from a single HSCT Centre were analyzed using flow cytometry. A statistical comparison between cell levels and clinical outcomes of HSCT was performed. Overall survival, acute graft-versus-host disease (aGVHD), chronic graft-versus-host disease (cGVHD), and relapse were used as the primary endpoints. Individual HSCT grafts showed considerable variation in their numbers of immune cell populations, including CD123+ dendritic cells and CD34+ cells, which may play a role in GVHD. Acute myeloid leukemia (AML) patients who developed aGVHD were transplanted with higher levels of effector CD3+ T, CD19+ B, and CD123+ dendritic cells than AML patients without aGVHD, whereas grafts with a high CD34+ content protected against aGVHD. AML patients with cGVHD had received grafts with a lower level of monocytes and a higher level of CD34+ cells than those without cGVHD. There is considerable variation in the levels of immune cell populations between HSCT grafts, and this variation is associated with outcomes of HSCT in AML patients. A detailed analysis of the immune cell content of the graft can be used in risk assessment of HSCT.
Collapse
Affiliation(s)
- Ulla Impola
- Finnish Red Cross Blood Service, Research and Development , Helsinki , Finland
| | - Antti Larjo
- Finnish Red Cross Blood Service, Research and Development , Helsinki , Finland
| | | | | | | | - Jukka Partanen
- Finnish Red Cross Blood Service, Research and Development , Helsinki , Finland
| |
Collapse
|
136
|
Cyclin-dependent kinase 5 activity is required for allogeneic T-cell responses after hematopoietic cell transplantation in mice. Blood 2016; 129:246-256. [PMID: 28064242 DOI: 10.1182/blood-2016-05-702738] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 10/16/2016] [Indexed: 01/14/2023] Open
Abstract
Molecular intermediates in T-cell activation pathways are crucial targets for the therapy and prevention of graft-versus-host disease (GVHD) following allogeneic hematopoietic cell transplantation (allo-HCT). We recently identified an essential role for cyclin-dependent kinase 5 (Cdk5) in T-cell activation and effector function, but the contribution of Cdk5 activity to the development of GVHD has not been explored. Using an established, preclinical, murine, GVHD model, we reveal that Cdk5 activity is increased in key target organs early after allo-HCT. We then generated chimeric mice (Cdk5+/+C or Cdk5-/-C) using hematopoietic progenitors from either embryonic day 16.5 Cdk5+/+ or Cdk5-/- embryos to enable analyses of the role of Cdk5 in GVHD, as germ line Cdk5 gene deletion is embryonically lethal. The immunophenotype of adult Cdk5-/-C mice is identical to control Cdk5+/+C mice. However, transplantation of donor Cdk5-/-C bone marrow and T cells dramatically reduced the severity of systemic and target organ GVHD. This phenotype is attributed to decreased T-cell migration to secondary lymphoid organs (SLOs), reduced in vivo proliferation within these organs, and fewer cytokine-producing donor T cells during GVHD development. Moreover, these defects in Cdk5-/- T-cell function are associated with altered CCR7 signaling following ligation by CCL19, a receptor:ligand interaction critical for T-cell migration into SLOs. Although Cdk5 activity in donor T cells contributed to graft-versus-tumor effects, pharmacologic inhibition of Cdk5 preserved leukemia-free survival. Collectively, our data implicate Cdk5 in allogeneic T-cell responses after HCT and as an important new target for therapeutic intervention.
Collapse
|
137
|
Efficacy of host-dendritic cell vaccinations with or without minor histocompatibility antigen loading, combined with donor lymphocyte infusion in multiple myeloma patients. Bone Marrow Transplant 2016; 52:228-237. [PMID: 27841858 DOI: 10.1038/bmt.2016.250] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 08/05/2016] [Accepted: 08/16/2016] [Indexed: 12/29/2022]
Abstract
Donor lymphocyte infusions (DLI) can induce durable remissions in multiple myeloma (MM) patients, but this occurs rather infrequently. As the graft-versus-tumor (GvT) effect of DLI depends on the presence of host-dendritic cells (DCs), we tested in a phase I/II trial whether the efficacy of DLI could be improved by simultaneous vaccination with host-DCs. We also analyzed the possibility of further improving the GvT effect by loading the DCs with peptides of mismatched hematopoietic cell-specific minor histocompatibility antigens (mHags). Fifteen MM patients not responding to a first DLI were included. Eleven patients could be treated with a second equivalent dose DLI combined with DC vaccinations, generated from host monocytes (moDC). For four patients, the DC products did not meet the quality criteria. In four of the treated patients the DCs were loaded with host mHag peptides. Toxicity was limited and no acute GvHD occurred. Most patients developed objective anti-host T-cell responses and in one patient a distinct mHag-specific T-cell response accompanied a temporary clinical response. These findings confirm that DLI combined with host-DC vaccination, either unloaded or loaded with mHag peptides, is feasible, safe and capable of inducing host-specific T-cell responses. The limited clinical effects may be improved by developing more immunogenic DC products or by combining this therapy with immune potentiating modalities like checkpoint inhibitors.
Collapse
|
138
|
Apostolova P, Zeiser R. The Role of Purine Metabolites as DAMPs in Acute Graft-versus-Host Disease. Front Immunol 2016; 7:439. [PMID: 27818661 PMCID: PMC5073102 DOI: 10.3389/fimmu.2016.00439] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 10/06/2016] [Indexed: 12/15/2022] Open
Abstract
Acute graft-versus-host disease (GvHD) causes high mortality in patients undergoing allogeneic hematopoietic cell transplantation. An early event in the classical pathogenesis of acute GvHD is tissue damage caused by the conditioning treatment or infection that consecutively leads to translocation of bacterial products [pathogen-associated molecular patterns (PAMPs)] into blood or lymphoid tissue, as well as danger-associated molecular patterns (DAMPs), mostly intracellular components that act as pro-inflammatory agents, once they are released into the extracellular space. A subtype of DAMPs is nucleotides, such as adenosine triphosphate released from dying cells that can activate the innate and adaptive immune system by binding to purinergic receptors. Binding to certain purinergic receptors leads to a pro-inflammatory microenvironment and promotes allogeneic T cell priming. After priming, T cells migrate to the acute GvHD target organs, mainly skin, liver, and the gastrointestinal tract and induce cell damage that further amplifies the release of intracellular components. This review summarizes the role of different purinergic receptors in particular P2X7 and P2Y2 as well as nucleotides in the pathogenesis of GvHD.
Collapse
Affiliation(s)
- Petya Apostolova
- Department of Hematology, Oncology and Stem Cell Transplantation, University Medical Center, Albert Ludwig University of Freiburg , Freiburg , Germany
| | - Robert Zeiser
- Department of Hematology, Oncology and Stem Cell Transplantation, University Medical Center, Albert Ludwig University of Freiburg , Freiburg , Germany
| |
Collapse
|
139
|
Haploidentical hematopoietic transplantation for the cure of leukemia: from its biology to clinical translation. Blood 2016; 128:2616-2623. [PMID: 27697774 DOI: 10.1182/blood-2016-07-730564] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 09/29/2016] [Indexed: 11/20/2022] Open
Abstract
The present review describes the biology of human leukocyte antigen haplotype mismatched ("haploidentical") transplantation, its translation to clinical practice to cure leukemia, and the results of current transplantation protocols. The 1990s saw what had been major drawbacks of haploidentical transplantation, ie, very strong host-versus-graft and graft-versus-host alloresponses, which led respectively to rejection and graft-versus-host disease (GVHD), being overcome through transplantation of a "mega-dose" of T cell-depleted peripheral blood hematopoietic progenitor cells and no posttransplant pharmacologic immunosuppression. The absence of posttransplant immunosuppression was an opportunity to discover natural killer cell alloreactions that eradicated acute myeloid leukemia and improved survival. Furthermore, it also unveiled the benefits of transplantation from mother donors, a likely consequence of the mother-to-child interaction during pregnancy. More recent transplantation protocols use unmanipulated (without ex vivo T-cell depletion) haploidentical grafts combined with enhanced posttransplant immunosuppression to help prevent GVHD. Unmanipulated grafts substantially extended the use of haploidentical transplantation with results than even rival those of matched hematopoietic transplantation. In T cell-depleted haploidentical transplantation, recent advances were made by the adoptive transfer of regulatory and conventional T cells.
Collapse
|
140
|
Zeiser R, Robson SC, Vaikunthanathan T, Dworak M, Burnstock G. Unlocking the Potential of Purinergic Signaling in Transplantation. Am J Transplant 2016; 16:2781-2794. [PMID: 27005321 PMCID: PMC5472988 DOI: 10.1111/ajt.13801] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/11/2016] [Accepted: 03/19/2016] [Indexed: 01/25/2023]
Abstract
Purinergic signaling has been recognized as playing an important role in inflammation, angiogenesis, malignancy, diabetes and neural transmission. Activation of signaling pathways downstream from purinergic receptors may also be implicated in transplantation and related vascular injury. Following transplantation, the proinflammatory "danger signal" adenosine triphosphate (ATP) is released from damaged cells and promotes proliferation and activation of a variety of immune cells. Targeting purinergic signaling pathways may promote immunosuppression and ameliorate inflammation. Under pathophysiological conditions, nucleotide-scavenging ectonucleotidases CD39 and CD73 hydrolyze ATP, ultimately, to the anti-inflammatory mediator adenosine. Adenosine suppresses proinflammatory cytokine production and is associated with improved graft survival and decreased severity of graft-versus-host disease. Furthermore, purinergic signaling is involved both directly and indirectly in the mechanism of action of several existing immunosuppressive drugs, such as calcineurin inhibitors and mammalian target of rapamycin inhibitors. Targeting of purinergic receptor pathways, particularly in the setting of combination therapies, could become a valuable immunosuppressive strategy in transplantation. This review focuses on the role of the purinergic signaling pathway in transplantation and immunosuppression and explores possible future applications in clinical practice.
Collapse
Affiliation(s)
- R. Zeiser
- Department of Hematology and Oncology, Freiburg University Medical Center, Albert-Ludwigs-University, Freiburg, Germany
| | - S. C. Robson
- Department of Medicine, Transplant Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA,Corresponding authors: Markus Dworak and Simon C. Robson, and
| | - T. Vaikunthanathan
- Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King’s College London, Great Maze Pond, London, UK
| | - M. Dworak
- Novartis Pharma, Nuernberg, Germany,Institute of Movement and Neurosciences, German Sport University Cologne, Cologne, Germany,Corresponding authors: Markus Dworak and Simon C. Robson, and
| | - G. Burnstock
- Autonomic Neuroscience Centre, University College Medical School, London, UK,Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
141
|
Effect of stem cell source on long-term chimerism and event-free survival in children with primary immunodeficiency disorders after fludarabine and melphalan conditioning regimen. J Allergy Clin Immunol 2016; 138:1152-1160. [DOI: 10.1016/j.jaci.2016.01.053] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 12/01/2015] [Accepted: 01/22/2016] [Indexed: 01/20/2023]
|
142
|
Boieri M, Shah P, Dressel R, Inngjerdingen M. The Role of Animal Models in the Study of Hematopoietic Stem Cell Transplantation and GvHD: A Historical Overview. Front Immunol 2016; 7:333. [PMID: 27625651 PMCID: PMC5003882 DOI: 10.3389/fimmu.2016.00333] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 08/18/2016] [Indexed: 12/13/2022] Open
Abstract
Bone marrow transplantation (BMT) is the only therapeutic option for many hematological malignancies, but its applicability is limited by life-threatening complications, such as graft-versus-host disease (GvHD). The last decades have seen great advances in the understanding of BMT and its related complications; in particular GvHD. Animal models are beneficial to study complex diseases, as they allow dissecting the contribution of single components in the development of the disease. Most of the current knowledge on the therapeutic mechanisms of BMT derives from studies in animal models. Parallel to BMT, the understanding of the pathophysiology of GvHD, as well as the development of new treatment regimens, has also been supported by studies in animal models. Pre-clinical experimentation is the basis for deep understanding and successful improvements of clinical applications. In this review, we retrace the history of BMT and GvHD by describing how the studies in animal models have paved the way to the many advances in the field. We also describe how animal models contributed to the understanding of GvHD pathophysiology and how they are fundamental for the discovery of new treatments.
Collapse
Affiliation(s)
- Margherita Boieri
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Pranali Shah
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen , Göttingen , Germany
| | - Ralf Dressel
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen , Göttingen , Germany
| | - Marit Inngjerdingen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
143
|
Tang B, Ren H, Liu H, Shi Y, Liu W, Dong Y, Yin Y, Miao S. CCR5 blockade combined with cyclosporine A attenuates liver GVHD by impairing T cells function. Inflamm Res 2016; 65:917-924. [PMID: 27423909 DOI: 10.1007/s00011-016-0974-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 05/26/2016] [Accepted: 07/09/2016] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVE Our preview study found that CCR5 blockade combined with cyclosporine A could attenuate the severity of liver GVHD. But the potential immunological mechanisms have not yet been explored. So our present study was designed to clarify the potential immunological mechanisms in mouse models after allo-HSCT. METHODS Firstly, we detected donor T cells homing to target organs, and analyzed the specific effector subsets in liver. Additionally, we assessed antigen-presenting cells (APCs), especially DCs and CD4+ T cells differentiation in secondary lymphoid organs. RESULTS Data showed that MVC combined with CsA reduced donor T cells migration to target organs in vivo. MVC and CsA treatment reduced the amount of donor T cells in the absolute numbers, also in donor CD4+ and CD8+ T cells by targeting at CCR5. And MVC co-injected with CsA was capable of slightly suppressing DC maturation, and reduced the percentage of Th1 and Th17 mainly by noncompetitive combination of CCR5. CONCLUSION Combined use of MVC and CsA was effective in attenuating liver GVHD in murine model. It can suppress DC maturation, affect T cells differentiation, and reduce donor T cells homing to target organs. This may offer a novel therapeutic perspective approach for clinical liver GVHD after allo-HSCT.
Collapse
Affiliation(s)
- Bo Tang
- Department of Hematology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Hanyun Ren
- Department of Hematology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing, 100034, China.
| | - Huihui Liu
- Department of Hematology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Yongjin Shi
- Department of Hematology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Wei Liu
- Department of Hematology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Yujun Dong
- Department of Hematology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Yue Yin
- Department of Hematology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Shengchao Miao
- Department of Hematology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| |
Collapse
|
144
|
MyD88 in donor bone marrow cells is critical for protection from acute intestinal graft-vs.-host disease. Mucosal Immunol 2016; 9:730-43. [PMID: 26442657 DOI: 10.1038/mi.2015.96] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 08/06/2015] [Indexed: 02/04/2023]
Abstract
To understand the role of myeloid differentiation factor 88 (MyD88) expressed by donor bone marrow (BM) in the pathophysiology of graft-vs.-host disease (GVHD), we investigated the effects of transplantation of MyD88-deficient T cell-depleted BM (MyD88KO TCD-BM) on the severity of GVHD. Transplantation with MyD88KO TCD-BM aggravated GVHD; serious gut damage was evident, with high infiltration of T cells into the intestines of recipients and markedly reduced expansion of CD11b(+)Gr-1(+) myeloid-derived suppressor cells (MDSCs). MDSCs from MyD88KO mice were defective in inducing donor T-cell apoptosis and inhibiting T-cell proliferation. Supplementation of transplanted mice with MDSCs from wild-type mice, but not MyD88KO mice, attenuated GVHD severity with reduced intestinal T-cell infiltration in MyD88KO TCD-BM recipients. Pretreatment of BM donors with lipopolysaccharide to increase MDSC levels and MyD88 transcription in the TCD-BM transplant alleviated GVHD severity and intestinal T-cell infiltration. The T cell/MDSC ratios were correlated with intestinal GVHD severity in both animal models and human patients. This study indicates that MyD88-dependent MDSC expansion from donor BM is critical for protection against fatal intestinal GVHD.
Collapse
|
145
|
Alloantigen presentation and graft-versus-host disease: fuel for the fire. Blood 2016; 127:2963-70. [PMID: 27030390 DOI: 10.1182/blood-2016-02-697250] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 03/05/2016] [Indexed: 12/16/2022] Open
Abstract
Allogeneic stem cell transplantation (SCT) is a unique procedure, primarily in patients with hematopoietic malignancies, involving chemoradiotherapy followed by the introduction of donor hematopoietic and immune cells into an inflamed and lymphopenic environment. Interruption of the process by which recipient alloantigen is presented to donor T cells to generate graft-versus-host disease (GVHD) represents an attractive therapeutic strategy to prevent morbidity and mortality after SCT and has been increasingly studied in the last 15 years. However, the immune activation resulting in GVHD has no physiological equivalent in nature; alloantigen is ubiquitous, persists indefinitely, and can be presented by multiple cell types at numerous sites, often on incompatible major histocompatibility complex, and occurs in the context of intense inflammation early after SCT. The recognition that alloantigen presentation is also critical to the development of immunological tolerance via both deletional and regulatory mechanisms further adds to this complexity. Finally, GVHD itself appears capable of inhibiting the presentation of microbiological antigens by donor dendritic cells late after SCT that is mandatory for the establishment of effective pathogen-specific immunity. Here, we review our current understanding of alloantigen, its presentation by various antigen-presenting cells, subsequent recognition by donor T cells, and the potential of therapeutic strategies interrupting this disease-initiating process to modify transplant outcome.
Collapse
|
146
|
Szyska M, Na IK. Bone Marrow GvHD after Allogeneic Hematopoietic Stem Cell Transplantation. Front Immunol 2016; 7:118. [PMID: 27066008 PMCID: PMC4811960 DOI: 10.3389/fimmu.2016.00118] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/15/2016] [Indexed: 12/15/2022] Open
Abstract
The bone marrow is the origin of all hematopoietic lineages and an important homing site for memory cells of the adaptive immune system. It has recently emerged as a graft-versus-host disease (GvHD) target organ after allogeneic stem cell transplantation (alloHSCT), marked by depletion of both hematopoietic progenitors and niche-forming cells. Serious effects on the restoration of hematopoietic function and immunological memory are common, especially in patients after myeloablative conditioning therapy. Cytopenia and durable immunodeficiency caused by the depletion of hematopoietic progenitors and destruction of bone marrow niches negatively influence the outcome of alloHSCT. The complex balance between immunosuppressive and cell-depleting treatments, GvHD and immune reconstitution, as well as the desirable graft-versus-tumor (GvT) effect remains a great challenge for clinicians.
Collapse
Affiliation(s)
- Martin Szyska
- Experimental and Clinical Research Center (ECRC) , Berlin , Germany
| | - Il-Kang Na
- Experimental and Clinical Research Center (ECRC), Berlin, Germany; Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
147
|
Magenau J, Runaas L, Reddy P. Advances in understanding the pathogenesis of graft-versus-host disease. Br J Haematol 2016; 173:190-205. [PMID: 27019012 DOI: 10.1111/bjh.13959] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 12/15/2015] [Indexed: 01/24/2023]
Abstract
Allogeneic haematopoietic stem cell transplantation (HCT) is a potent immunotherapy with curative potential for several haematological disorders. Overcoming the immunological barrier of acute graft-versus-host disease (GVHD) remains a fundamental impediment to expanding the efficacy of HCT. GVHD reflects a complex pathological interaction between the innate and adaptive immune systems of the host and donor. Over the past decade there has been a tremendous advancement in our understanding of the cellular and molecular underpinnings of this devastating disease. In this review, we cover several recently appreciated facets of GVHD pathogenesis including novel extracellular mediators of inflammation, immune subsets, intracellular signal transduction, post-translation modifications and epigenetic regulation. We begin to develop general themes regarding the immunological pathways in GVHD pathogenesis, discuss critical outstanding questions, and explore new avenues for GVHD treatment and prevention.
Collapse
Affiliation(s)
- John Magenau
- Blood and Marrow Transplant Program, Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Lyndsey Runaas
- Blood and Marrow Transplant Program, Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Pavan Reddy
- Blood and Marrow Transplant Program, Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
148
|
Villa NY, Rahman MM, McFadden G, Cogle CR. Therapeutics for Graft-versus-Host Disease: From Conventional Therapies to Novel Virotherapeutic Strategies. Viruses 2016; 8:85. [PMID: 27011200 PMCID: PMC4810275 DOI: 10.3390/v8030085] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 03/09/2016] [Accepted: 03/09/2016] [Indexed: 02/06/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) has a curative potential for many hematologic malignancies and blood diseases. However, the success of allo-HSCT is limited by graft-versus-host disease (GVHD), an immunological syndrome that involves inflammation and tissue damage mediated by donor lymphocytes. Despite immune suppression, GVHD is highly incident even after allo-HSCT using human leukocyte antigen (HLA)-matched donors. Therefore, alternative and more effective therapies are needed to prevent or control GVHD while preserving the beneficial graft-versus-cancer (GVC) effects against residual disease. Among novel therapeutics for GVHD, oncolytic viruses such as myxoma virus (MYXV) are receiving increased attention due to their dual role in controlling GVHD while preserving or augmenting GVC. This review focuses on the molecular basis of GVHD, as well as state-of-the-art advances in developing novel therapies to prevent or control GVHD while minimizing impact on GVC. Recent literature regarding conventional and the emerging therapies are summarized, with special emphasis on virotherapy to prevent GVHD. Recent advances using preclinical models with oncolytic viruses such as MYXV to ameliorate the deleterious consequences of GVHD, while maintaining or improving the anti-cancer benefits of GVC will be reviewed.
Collapse
Affiliation(s)
- Nancy Y Villa
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL 32610, USA.
| | - Masmudur M Rahman
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA.
| | - Grant McFadden
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA.
| | - Christopher R Cogle
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
149
|
Seldon TA, Pryor R, Palkova A, Jones ML, Verma ND, Findova M, Braet K, Sheng Y, Fan Y, Zhou EY, Marks JD, Munro T, Mahler SM, Barnard RT, Fromm PD, Silveira PA, Elgundi Z, Ju X, Clark GJ, Bradstock KF, Munster DJ, Hart DNJ. Immunosuppressive human anti-CD83 monoclonal antibody depletion of activated dendritic cells in transplantation. Leukemia 2016; 30:692-700. [PMID: 26286117 DOI: 10.1038/leu.2015.231] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 07/27/2015] [Indexed: 02/05/2023]
Abstract
Current immunosuppressive/anti-inflammatory agents target the responding effector arm of the immune response and their nonspecific action increases the risk of infection and malignancy. These effects impact on their use in allogeneic haematopoietic cell transplantation and other forms of transplantation. Interventions that target activated dendritic cells (DCs) have the potential to suppress the induction of undesired immune responses (for example, graft versus host disease (GVHD) or transplant rejection) and to leave protective T-cell immune responses intact (for example, cytomegalovirus (CMV) immunity). We developed a human IgG1 monoclonal antibody (mAb), 3C12, specific for CD83, which is expressed on activated but not resting DC. The 3C12 mAb and an affinity improved version, 3C12C, depleted CD83(+) cells by CD16(+) NK cell-mediated antibody-dependent cellular cytotoxicity, and inhibited allogeneic T-cell proliferation in vitro. A single dose of 3C12C prevented human peripheral blood mononuclear cell-induced acute GVHD in SCID mouse recipients. The mAb 3C12C depleted CMRF-44(+)CD83(bright) activated DC but spared CD83(dim/-) DC in vivo. It reduced human T-cell activation in vivo and maintained the proportion of CD4(+) FoxP3(+) CD25(+) Treg cells and also viral-specific CD8(+) T cells. The anti-CD83 mAb, 3C12C, merits further evaluation as a new immunosuppressive agent in transplantation.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Antigens, CD/genetics
- Antigens, CD/immunology
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Cell Proliferation/drug effects
- Cytotoxicity, Immunologic/drug effects
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/pathology
- Female
- Gene Expression
- Graft Rejection/immunology
- Graft Rejection/mortality
- Graft Rejection/pathology
- Graft Rejection/prevention & control
- Graft vs Host Disease/immunology
- Graft vs Host Disease/mortality
- Graft vs Host Disease/pathology
- Graft vs Host Disease/prevention & control
- Humans
- Immunoglobulins/genetics
- Immunoglobulins/immunology
- Immunosuppressive Agents/pharmacology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/pathology
- Leukocytes, Mononuclear/cytology
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/transplantation
- Membrane Glycoproteins/antagonists & inhibitors
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Mice
- Mice, SCID
- Survival Analysis
- Transplantation, Heterologous
- CD83 Antigen
Collapse
Affiliation(s)
- T A Seldon
- DC Program, Mater Medical Research Institute, Brisbane, Queensland, Australia
- Co-operative Research Centre for Biomarker Translation, Melbourne, Victoria, Australia
| | - R Pryor
- DC Program, Mater Medical Research Institute, Brisbane, Queensland, Australia
| | - A Palkova
- DC Program, Mater Medical Research Institute, Brisbane, Queensland, Australia
| | - M L Jones
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, Australia
| | - N D Verma
- Dendritic Cell Research, ANZAC Research Institute, Concord, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - M Findova
- DC Program, Mater Medical Research Institute, Brisbane, Queensland, Australia
| | - K Braet
- DC Program, Mater Medical Research Institute, Brisbane, Queensland, Australia
| | - Y Sheng
- DC Program, Mater Medical Research Institute, Brisbane, Queensland, Australia
- Co-operative Research Centre for Biomarker Translation, Melbourne, Victoria, Australia
| | - Y Fan
- Anesthesia, Helen Diller Family Comprehensive Cancer Centre, University of California, San Francisco, CA, USA
| | - E Y Zhou
- Anesthesia, Helen Diller Family Comprehensive Cancer Centre, University of California, San Francisco, CA, USA
| | - J D Marks
- Anesthesia, Helen Diller Family Comprehensive Cancer Centre, University of California, San Francisco, CA, USA
| | - T Munro
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, Australia
| | - S M Mahler
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, Australia
| | - R T Barnard
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - P D Fromm
- Dendritic Cell Research, ANZAC Research Institute, Concord, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - P A Silveira
- Dendritic Cell Research, ANZAC Research Institute, Concord, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Z Elgundi
- Dendritic Cell Research, ANZAC Research Institute, Concord, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - X Ju
- Dendritic Cell Research, ANZAC Research Institute, Concord, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - G J Clark
- DC Program, Mater Medical Research Institute, Brisbane, Queensland, Australia
- Co-operative Research Centre for Biomarker Translation, Melbourne, Victoria, Australia
- Dendritic Cell Research, ANZAC Research Institute, Concord, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - K F Bradstock
- Dendritic Cell Research, ANZAC Research Institute, Concord, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - D J Munster
- DC Program, Mater Medical Research Institute, Brisbane, Queensland, Australia
- Co-operative Research Centre for Biomarker Translation, Melbourne, Victoria, Australia
| | - D N J Hart
- DC Program, Mater Medical Research Institute, Brisbane, Queensland, Australia
- Co-operative Research Centre for Biomarker Translation, Melbourne, Victoria, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, Australia
- Dendritic Cell Research, ANZAC Research Institute, Concord, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
150
|
Fu J, Wu Y, Nguyen H, Heinrichs J, Schutt S, Liu Y, Liu C, Jin J, Anasetti C, Yu XZ. T-bet Promotes Acute Graft-versus-Host Disease by Regulating Recipient Hematopoietic Cells in Mice. THE JOURNAL OF IMMUNOLOGY 2016; 196:3168-79. [PMID: 26903480 DOI: 10.4049/jimmunol.1501020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 01/21/2016] [Indexed: 01/16/2023]
Abstract
Beyond its critical role in T cells, T-bet regulates the functions of APCs including dendritic cells and B cells, as well as NK cells. Given that recipient APCs are essential for priming allogeneic T cells and recipient NK or T cells are able to reject allogeneic donor cells, we evaluated the role of T-bet on the host in acute graft-versus-host disease (GVHD) using murine models of allogeneic bone marrow transplantation. T-bet(-/-) recipients developed significantly milder GVHD than their wild type counterparts in MHC-mismatched or CD4-dependent minor histocompatibility Ag-mismatched models. Allogeneic donor T cells, in particular, CD4 subset, significantly reduced IFN-γ production, proliferation and migration, and caused less injury in liver and gut of T-bet(-/-) recipients. We further observed that T-bet on recipient hematopoietic cells was primarily responsible for the donor T cell response and pathogenicity in GVHD. T-bet(-/-) dendritic cells expressed higher levels of Trail, whereas they produced lower levels of IFN-γ and IL-12/23 p40, as well as chemokine CXCL9, resulting in significantly higher levels of apoptosis, less priming, and infiltration of donor T cells. Meanwhile, NK cells in T-bet(-/-) hosts partially contribute to the decreased donor T cell proliferation. Furthermore, although T-bet on hematopoietic cells was required for GVHD development, it was largely dispensable for the graft-versus-leukemia effect. Taken together with our previous findings, we propose that T-bet is a potential therapeutic target for the control of GVHD through regulating donor T cells and recipient hematopoietic cells.
Collapse
Affiliation(s)
- Jianing Fu
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL 33612; Department of Immunology, Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612; Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425
| | - Yongxia Wu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425
| | - Hung Nguyen
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425
| | - Jessica Heinrichs
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425
| | - Steven Schutt
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425
| | - Yuejun Liu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425
| | - Chen Liu
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611
| | - Junfei Jin
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425; Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001 Guangxi, China; and
| | - Claudio Anasetti
- Department of Immunology, Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | - Xue-Zhong Yu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425; Department of Medicine, Medical University of South Carolina, Charleston, SC 29425
| |
Collapse
|