1
|
Zhu J, Yang L, Xia J, Zhou N, Zhu J, Zhu H, Chen J, Qing K, Duan CW. Interleukin-27 Promotes the Generation of Myeloid-derived Suppressor Cells to Alleviate Graft-versus-host Disease. Transplantation 2024:00007890-990000000-00771. [PMID: 38773837 DOI: 10.1097/tp.0000000000005069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2024]
Abstract
BACKGROUND Stimulation of myeloid-derived suppressor cell (MDSC) formation represents a potential curative therapeutic approach for graft-versus-host disease (GVHD), which significantly impacts the prognosis of allogeneic hematopoietic stem cell transplantation. However, the lack of an effective strategy for inducing MDSC production in vivo has hindered their clinical application. In our previous study, MDSC expansion was observed in interleukin (IL)-27-treated mice. METHODS In this study, we overexpressed exogenous IL-27 in mice using a recombinant adeno-associated virus vector to investigate its therapeutic and exacerbating effects in murine GVHD models. RESULTS In our study, we demonstrated that exogenous administration of IL-27 significantly suppressed GVHD development in a mouse model. We found that IL-27 treatment indirectly inhibited the proliferation and activation of donor T cells by rapidly expanding recipient and donor myeloid cells, which act as MDSCs after irradiation or under inflammatory conditions, rather than through regulatory T-cell expansion. Additionally, IL-27 stimulated MDSC expansion by enhancing granulocyte-monocyte progenitor generation. Notably, we verified that IL-27 signaling in donor T cells exerted an antagonistic effect on GVHD prevention and treatment. Further investigation revealed that combination therapy involving IL-27 and T-cell depletion exhibited remarkable preventive effects on GVHD in both mouse and xenogeneic GVHD models. CONCLUSIONS Collectively, these findings suggest that IL-27 promotes MDSC generation to reduce the incidence of GVHD, whereas targeted activation of IL-27 signaling in myeloid progenitors or its combination with T-cell depletion represents a potential strategy for GVHD therapy.
Collapse
Affiliation(s)
- Jianmin Zhu
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liting Yang
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Xia
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Neng Zhou
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiayao Zhu
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Zhu
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Chen
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kai Qing
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cai-Wen Duan
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Yuan X, Jiang H, Fu D, Rech JC, Robida A, Rajanayake K, Yuan H, He M, Wen B, Sun D, Liu C, Chinnaswamy K, Stuckey JA, Paczesny S, Yang CY. Prophylactic Mitigation of Acute Graft versus Host Disease by Novel 2-(Pyrrolidin-1-ylmethyl)pyrrole-Based Stimulation-2 (ST2) Inhibitors. ACS Pharmacol Transl Sci 2023; 6:1275-1287. [PMID: 37705593 PMCID: PMC10496145 DOI: 10.1021/acsptsci.3c00122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Indexed: 09/15/2023]
Abstract
Hematopoietic cell transplantation (HCT) is a proven and potentially curable therapy for hematological malignancies and inherited hematological disease. The main risk of HCT is the development of graft versus host disease (GVHD) acquired in up to 50% of patients. Upregulation of soluble ST2 (sST2) is a key clinical biomarker for GVHD prognosis and was shown to be a potential therapeutic target for GVHD. Agents targeting sST2 to reduce the sST2 level after HCT have the potential to mitigate GVHD progression. Here, we report 32 (or XY52) as the lead ST2 inhibitor from our optimization campaign. XY52 had improved inhibitory activity and metabolic stability in vitro and in vivo. XY52 suppressed proinflammatory T-cell proliferation while increasing regulatory T cells in vitro. In a clinically relevant GVHD model, a 21-day prophylactic regimen of XY52 reduced plasma sST2 and IFN-γ levels and GVHD score and extended survival in mice. XY52 represented a significant improvement over our previous compound, iST2-1, and further optimization of XY52 is warranted. The small-molecule ST2 inhibitors can potentially be used as a biomarker-guided therapy for mitigating GVHD in future clinical applications.
Collapse
Affiliation(s)
- Xinrui Yuan
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Hua Jiang
- Department
of Microbiology & Immunology, Medical
University of South Carolina, Charleston, South Carolina 29425-2503, United States
| | - Denggang Fu
- Department
of Microbiology & Immunology, Medical
University of South Carolina, Charleston, South Carolina 29425-2503, United States
| | - Jason C. Rech
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Aaron Robida
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Krishani Rajanayake
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Hebao Yuan
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Miao He
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Bo Wen
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Duxin Sun
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Chen Liu
- Department
of Pathology, Yale University, New Haven, Connecticut 06520, United States
| | - Krishnapriya Chinnaswamy
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jeanne A. Stuckey
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Sophie Paczesny
- Department
of Microbiology & Immunology, Medical
University of South Carolina, Charleston, South Carolina 29425-2503, United States
| | - Chao-Yie Yang
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| |
Collapse
|
3
|
Li J, Wang P, Zhou T, Jiang W, Wu H, Zhang S, Deng L, Wang H. Neuroprotective effects of interleukin 10 in spinal cord injury. Front Mol Neurosci 2023; 16:1214294. [PMID: 37492521 PMCID: PMC10363608 DOI: 10.3389/fnmol.2023.1214294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 06/26/2023] [Indexed: 07/27/2023] Open
Abstract
Spinal cord injury (SCI) starts with a mechanical and/or bio-chemical insult, followed by a secondary phase, leading progressively to severe collapse of the nerve tissue. Compared to the peripheral nervous system, injured spinal cord is characterized by weak axonal regeneration, which leaves most patients impaired or paralyzed throughout lifetime. Therefore, confining, alleviating, or reducing the expansion of secondary injuries and promoting functional connections between rostral and caudal regions of lesion are the main goals of SCI therapy. Interleukin 10 (IL-10), as a pivotal anti-inflammatory and immunomodulatory cytokine, exerts a wide spectrum of positive effects in the treatment of SCI. The mechanisms underlying therapeutic effects mainly include anti-oxidative stress, limiting excessive inflammation, anti-apoptosis, antinociceptive effects, etc. Furthermore, IL-10 displays synergistic effects when combined with cell transplantation or neurotrophic factor, enhancing treatment outcomes. This review lists pleiotropic mechanisms underlying IL-10-mediated neuroprotection after SCI, which may offer fresh perspectives for clinical translation.
Collapse
Affiliation(s)
- Juan Li
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| | - Pei Wang
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| | - Ting Zhou
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| | - Wenwen Jiang
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| | - Hang Wu
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| | - Shengqi Zhang
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| | - Lingxiao Deng
- Department of Neurological Surgery, Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Hongxing Wang
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| |
Collapse
|
4
|
Zheng YY, Yang XT, Lin GQ, Bian MR, Si YJ, Zhang XX, Zhang YM, Wu DP. [Clinical study of 19 cases of steroid-refractory gastrointestinal acute graft-versus-host disease after allogeneic hematopoietic stem cell transplantation with fecal microbiota transplantation]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2023; 44:401-407. [PMID: 37550190 PMCID: PMC10440624 DOI: 10.3760/cma.j.issn.0253-2727.2023.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Indexed: 08/09/2023]
Abstract
Objective: To investigate the clinical efficacy of fecal microbiota transplantation (FMT) for treating steroid-refractory gastrointestinal acute graft-versus-host disease (GI-aGVHD) . Methods: This analysis included 29 patients with hematology who developed steroid-refractory GI-aGVHD after allogeneic hematopoietic stem cell transplantation (allo-HSCT) in Huaian Hospital Affiliated to Xuzhou Medical University from March 2017 to March 2022. Among them, 19 patients underwent FMT treatment (the FMT group) and 10 patients did not (the control group). The efficacy and safety of FMT were assessed, as well as the changes in intestinal microbiota abundance, lymphocyte subpopulation ratio, peripheral blood inflammatory cytokines, and GVHD biomarkers before and after FMT treatment. Results: ① Complete remission of clinical symptoms after FMT was achieved by 13 (68.4%) patients and 2 (20.0%) controls, with a statistically significant difference (P<0.05). Intestinal microbiota diversity increased and gradually recovered to normal levels after FMT and FMT-related infections did not occur. ②The proportion of CD3(+) and CD8(+) cells in the FMT group after treatment decreased compared with the control group, and the ratio of CD4(+), regulatory T cells (Treg), and CD4(+)/CD8(+) cells increased (all P< 0.05). The interleukin (IL) -6 concentration in the FMT group was lower than that in the control group [4.15 (1.91-5.71) ng/L vs 6.82 (2.40-8.91) ng/L, P=0.040], and the IL-10 concentration in the FMT group was higher than that in the control group [12.11 (5.69-20.36) ng/L vs 7.51 (4.10-9.58) ng/L, P=0.024]. Islet-derived protein 3α (REG3α) was significantly increased in patients with GI-aGVHD, and the REG3α level in the FMT group was lower than that in the control group after treatment [30.70 (10.50-105.00) μg/L vs 74.35 (33.50-139.50) μg/L, P=0.021]. Conclusion: FMT is a safe and effective method for the treatment of steroid-refractory GI-aGVHD by restoring intestinal microbiota diversity, regulating inflammatory cytokines, and upregulating Treg cells.
Collapse
Affiliation(s)
- Y Y Zheng
- Department of Hematology, Huai'an Hospital Affiliated to Xuzhou Medical Universitity, Huai'an 223002, China
| | - X T Yang
- Department of Hematology, Huai'an Hospital Affiliated to Xuzhou Medical Universitity, Huai'an 223002, China
| | - G Q Lin
- Department of Hematology, Huai'an Hospital Affiliated to Xuzhou Medical Universitity, Huai'an 223002, China
| | - M R Bian
- Department of Hematology, Huai'an Hospital Affiliated to Xuzhou Medical Universitity, Huai'an 223002, China
| | - Y J Si
- Department of Hematology, Huai'an Hospital Affiliated to Xuzhou Medical Universitity, Huai'an 223002, China
| | - X X Zhang
- Department of Hematology, Huai'an Hospital Affiliated to Xuzhou Medical Universitity, Huai'an 223002, China
| | - Y M Zhang
- Department of Hematology, Huai'an Hospital Affiliated to Xuzhou Medical Universitity, Huai'an 223002, China
| | - D P Wu
- Department of Hematology, the First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Disease, Suzhou 215006, China
| |
Collapse
|
5
|
Niebergall-Roth E, Frank NY, Ganss C, Frank MH, Kluth MA. Skin-Derived ABCB5 + Mesenchymal Stem Cells for High-Medical-Need Inflammatory Diseases: From Discovery to Entering Clinical Routine. Int J Mol Sci 2022; 24:66. [PMID: 36613507 PMCID: PMC9820160 DOI: 10.3390/ijms24010066] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/16/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
The ATP-binding cassette superfamily member ABCB5 identifies a subset of skin-resident mesenchymal stem cells (MSCs) that exhibit potent immunomodulatory and wound healing-promoting capacities along with superior homing ability. The ABCB5+ MSCs can be easily accessed from discarded skin samples, expanded, and delivered as a highly homogenous medicinal product with standardized potency. A range of preclinical studies has suggested therapeutic efficacy of ABCB5+ MSCs in a variety of currently uncurable skin and non-skin inflammatory diseases, which has been substantiated thus far by distinct clinical trials in chronic skin wounds or recessive dystrophic epidermolysis bullosa. Therefore, skin-derived ABCB5+ MSCs have the potential to provide a breakthrough at the forefront of MSC-based therapies striving to fulfill current unmet medical needs. The most recent milestones in this regard are the approval of a phase III pivotal trial of ABCB5+ MSCs for treatment of recessive dystrophic and junctional epidermolysis bullosa by the US Food and Drug Administration, and national market access of ABCB5+ MSCs (AMESANAR®) for therapy-refractory chronic venous ulcers under the national hospital exemption pathway in Germany.
Collapse
Affiliation(s)
| | - Natasha Y. Frank
- Department of Medicine, VA Boston Healthcare System, Boston, MA 02132, USA
- Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
- Transplant Research Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Christoph Ganss
- TICEBA GmbH, 69120 Heidelberg, Germany
- RHEACELL GmbH & Co. KG, 69120 Heidelberg, Germany
| | - Markus H. Frank
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
- Transplant Research Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Dermatology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- School of Medical and Health Sciences, Edith Cowan University, Perth 6027, Australia
| | - Mark A. Kluth
- TICEBA GmbH, 69120 Heidelberg, Germany
- RHEACELL GmbH & Co. KG, 69120 Heidelberg, Germany
| |
Collapse
|
6
|
Challenges and opportunities targeting mechanisms of epithelial injury and recovery in acute intestinal graft-versus-host disease. Mucosal Immunol 2022; 15:605-619. [PMID: 35654837 PMCID: PMC9259481 DOI: 10.1038/s41385-022-00527-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/21/2022] [Accepted: 05/16/2022] [Indexed: 02/04/2023]
Abstract
Despite advances in immunosuppressive prophylaxis and overall supportive care, gastrointestinal (GI) graft-versus-host disease (GVHD) remains a major, lethal side effect after allogeneic hematopoietic stem cell transplantation (allo-HSCT). It has become increasingly clear that the intestinal epithelium, in addition to being a target of transplant-related toxicity and GVHD, plays an important role in the onset of GVHD. Over the last two decades, increased understanding of the epithelial constituents and their microenvironment has led to the development of novel prophylactic and therapeutic interventions, with the potential to protect the intestinal epithelium from GVHD-associated damage and promote its recovery following insult. In this review, we will discuss intestinal epithelial injury and the role of the intestinal epithelium in GVHD pathogenesis. In addition, we will highlight possible approaches to protect the GI tract from damage posttransplant and to stimulate epithelial regeneration, in order to promote intestinal recovery. Combined treatment modalities integrating immunomodulation, epithelial protection, and induction of regeneration may hold the key to unlocking mucosal recovery and optimizing therapy for acute intestinal GVHD.
Collapse
|
7
|
Wang S, Wang D, Chang Y, Geng L, Qiang P, Sun G, Tang B, Zhao X, Zhou Z, Liu H. Elevated RAP1A expression correlates with the severity of acute GVHD after umbilical cord blood transplantation. Transpl Immunol 2022; 71:101546. [DOI: 10.1016/j.trim.2022.101546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/15/2022] [Accepted: 01/25/2022] [Indexed: 12/24/2022]
|
8
|
Hill GR, Koyama M. Cytokines and costimulation in acute graft-versus-host disease. Blood 2020; 136:418-428. [PMID: 32526028 PMCID: PMC7378458 DOI: 10.1182/blood.2019000952] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/18/2020] [Indexed: 12/11/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (alloSCT) is an important curative therapy for high-risk hematological malignancies, but the development of severe and/or steroid-refractory acute graft-versus-host disease (aGVHD) remains a significant limitation to optimal outcomes. New approaches to prevent and treat aGVHD remain an unmet need that can be best addressed by understanding the complex disease pathophysiology. It is now clear that chemoradiotherapy used prior to alloSCT induces the release of endogenous alarmins (eg, HMGB-1, ATP, IL-1α, IL-33) from recipient tissue. Exogenous pathogen-derived molecules (eg, lipopolysaccharide, nucleic acids) also translocate from the gastrointestinal tract lumen. Together, these danger signals activate antigen-presenting cells (APCs) to efficiently present alloantigen to donor T cells while releasing cytokines (eg, interleukin-12 [IL-12], IL-23, IL-6, IL-27, IL-10, transforming growth factor-β) that expand and differentiate both pathogenic and regulatory donor T cells. Concurrent costimulatory signals at the APC-T-cell interface (eg, CD80/CD86-CD28, CD40-CD40L, OX40L-OX40, CD155/CD112-DNAM-1) and subsequent coinhibitory signals (eg, CD80/CD86-CTLA4, PDL1/2-PD1, CD155/CD112-TIGIT) are critical to the acquisition of effector T-cell function and ensuing secretion of pathogenic cytokines (eg, IL-17, interferon-γ, tissue necrosis factor, granulocyte-macrophage colony-stimulating factor) and cytolytic degranulation pathway effectors (eg, perforin/granzyme). This review focuses on the combination of cytokine and costimulatory networks at the T-cell surface that culminates in effector function and subsequent aGVHD in target tissue. Together, these pathways now represent robust and clinically tractable targets for preventing the initiation of deleterious immunity after alloSCT.
Collapse
Affiliation(s)
- Geoffrey R Hill
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA; and
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Motoko Koyama
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA; and
| |
Collapse
|
9
|
Talvard-Balland N, Sutra Del Galy A, Michonneau D, Le Buanec H, Chasset F, Robin M, Peffault de Latour R, Xhaard A, Sicre de Fontbrune F, Parquet N, Duchez S, Schiavon V, Rybojad M, Bergeron-Lafaurie A, Bagot M, Bensussan A, Caillat-Zucman S, Socié G, Bouaziz JD, de Masson A. Expansion of Circulating CD49b +LAG3 + Type 1 Regulatory T Cells in Human Chronic Graft-Versus-Host Disease. J Invest Dermatol 2020; 141:193-197.e2. [PMID: 32428542 DOI: 10.1016/j.jid.2020.04.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/27/2020] [Accepted: 04/24/2020] [Indexed: 12/21/2022]
Affiliation(s)
- Nana Talvard-Balland
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France; Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France
| | - Aurélien Sutra Del Galy
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France; Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France; Service d'Hématologie-Greffes, AP-HP, Hôpital Saint-Louis, Paris, France
| | - David Michonneau
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France; Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France; Service d'Hématologie-Greffes, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Helene Le Buanec
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France; Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France
| | - Francois Chasset
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France; Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France; Service de Dermatologie, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Marie Robin
- Service d'Hématologie-Greffes, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Régis Peffault de Latour
- Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France; Service d'Hématologie-Greffes, AP-HP, Hôpital Saint-Louis, Paris, France; INSERM UMRS 1160, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France
| | - Alienor Xhaard
- Service d'Hématologie-Greffes, AP-HP, Hôpital Saint-Louis, Paris, France
| | | | - Nathalie Parquet
- Service d'Aphérèse Thérapeutique, AP-HP, Hôpital Saint-Louis, France
| | - Sophie Duchez
- UMR CNRS 7212, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France
| | - Valérie Schiavon
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France
| | - Michel Rybojad
- Service de Dermatologie, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Anne Bergeron-Lafaurie
- Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France; Service de Pneumologie, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Martine Bagot
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France; Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France; Service de Dermatologie, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Armand Bensussan
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France; Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France
| | - Sophie Caillat-Zucman
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France; Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France; Laboratoire d'Immunologie, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Gérard Socié
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France; Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France; Service d'Hématologie-Greffes, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Jean-David Bouaziz
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France; Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France; Service de Dermatologie, AP-HP, Hôpital Saint-Louis, Paris, France.
| | - Adèle de Masson
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France; Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France; Service de Dermatologie, AP-HP, Hôpital Saint-Louis, Paris, France
| |
Collapse
|
10
|
Stokes J, Hoffman EA, Molina MS, Kummet N, Simpson RJ, Zeng Y, Katsanis E. Bendamustine with total body irradiation conditioning yields tolerant T-cells while preserving T-cell-dependent graft-versus-leukemia. Oncoimmunology 2020; 9:1758011. [PMID: 32391190 PMCID: PMC7199810 DOI: 10.1080/2162402x.2020.1758011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/01/2020] [Accepted: 04/15/2020] [Indexed: 12/16/2022] Open
Abstract
Graft-versus-host disease (GvHD) remains a significant impediment to allogeneic hematopoietic cell transplantation (HCT) success, necessitating studies focused on alleviating GvHD, while preserving the graft-versus-leukemia (GvL) effect. Based on our previous studies showing bendamustine with total body irradiation (BEN-TBI) conditioning reduces GvHD compared to the current clinical standard of care cyclophosphamide (CY)-TBI in a murine MHC-mismatched bone marrow transplantation (BMT) model, this study aimed to evaluate the role and fate of donor T-cells following BEN-TBI conditioning. We demonstrate that BEN-TBI reduces GvHD compared to CY-TBI independently of T regulatory cells (Tregs). BEN-TBI conditioned mice have a smaller proportion and less activated donor T-cells, with lower CD47 expression, early post-transplant, but no sustained phenotypic differences in T-cells. In BEN-TBI conditioned mice, donor T-cells gain tolerance specific to host MHC antigens. Though these T-cells are tolerant to host antigens, we demonstrate that BEN-TBI preserves a T-cell-dependent GvL effect. These findings indicate that BEN-TBI conditioning reduces GvHD without compromising GvL, warranting its further investigation as a potentially safer and more efficacious clinical alternative to CY-TBI.
Collapse
Affiliation(s)
- Jessica Stokes
- Department of Pediatrics, University of Arizona, Tucson, AZ, USA
| | - Emely A Hoffman
- Department of Pediatrics, University of Arizona, Tucson, AZ, USA
| | - Megan S Molina
- Department of Pediatrics, University of Arizona, Tucson, AZ, USA.,Department of Immunobiology, University of Arizona, Tucson, AZ, USA
| | - Nicole Kummet
- Department of Pediatrics, University of Arizona, Tucson, AZ, USA.,Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ, USA
| | - Richard J Simpson
- Department of Pediatrics, University of Arizona, Tucson, AZ, USA.,Department of Immunobiology, University of Arizona, Tucson, AZ, USA.,Department of Nutritional Sciences, University of Arizona, Tucson, AZ, USA.,The University of Arizona Cancer Center, Tucson, AZ, USA
| | - Yi Zeng
- Department of Pediatrics, University of Arizona, Tucson, AZ, USA.,The University of Arizona Cancer Center, Tucson, AZ, USA
| | - Emmanuel Katsanis
- Department of Pediatrics, University of Arizona, Tucson, AZ, USA.,Department of Immunobiology, University of Arizona, Tucson, AZ, USA.,The University of Arizona Cancer Center, Tucson, AZ, USA.,Department of Medicine, University of Arizona, Tucson, AZ, USA.,Department of Pathology, University of Arizona, Tucson, AZ, USA
| |
Collapse
|