1
|
Li MM, Huang Y, Sumathipala M, Liang MQ, Valdeolivas A, Ananthakrishnan AN, Liao K, Marbach D, Zitnik M. Contextual AI models for single-cell protein biology. Nat Methods 2024; 21:1546-1557. [PMID: 39039335 PMCID: PMC11310085 DOI: 10.1038/s41592-024-02341-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 06/10/2024] [Indexed: 07/24/2024]
Abstract
Understanding protein function and developing molecular therapies require deciphering the cell types in which proteins act as well as the interactions between proteins. However, modeling protein interactions across biological contexts remains challenging for existing algorithms. Here we introduce PINNACLE, a geometric deep learning approach that generates context-aware protein representations. Leveraging a multiorgan single-cell atlas, PINNACLE learns on contextualized protein interaction networks to produce 394,760 protein representations from 156 cell type contexts across 24 tissues. PINNACLE's embedding space reflects cellular and tissue organization, enabling zero-shot retrieval of the tissue hierarchy. Pretrained protein representations can be adapted for downstream tasks: enhancing 3D structure-based representations for resolving immuno-oncological protein interactions, and investigating drugs' effects across cell types. PINNACLE outperforms state-of-the-art models in nominating therapeutic targets for rheumatoid arthritis and inflammatory bowel diseases and pinpoints cell type contexts with higher predictive capability than context-free models. PINNACLE's ability to adjust its outputs on the basis of the context in which it operates paves the way for large-scale context-specific predictions in biology.
Collapse
Affiliation(s)
- Michelle M Li
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Yepeng Huang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Marissa Sumathipala
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Man Qing Liang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Alberto Valdeolivas
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Ashwin N Ananthakrishnan
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Katherine Liao
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Daniel Marbach
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Marinka Zitnik
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, Allston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Harvard Data Science Initiative, Cambridge, MA, USA.
| |
Collapse
|
2
|
Li MM, Huang Y, Sumathipala M, Liang MQ, Valdeolivas A, Ananthakrishnan AN, Liao K, Marbach D, Zitnik M. Contextual AI models for single-cell protein biology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.18.549602. [PMID: 37503080 PMCID: PMC10370131 DOI: 10.1101/2023.07.18.549602] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Understanding protein function and developing molecular therapies require deciphering the cell types in which proteins act as well as the interactions between proteins. However, modeling protein interactions across biological contexts remains challenging for existing algorithms. Here, we introduce Pinnacle, a geometric deep learning approach that generates context-aware protein representations. Leveraging a multi-organ single-cell atlas, Pinnacle learns on contextualized protein interaction networks to produce 394,760 protein representations from 156 cell type contexts across 24 tissues. Pinnacle's embedding space reflects cellular and tissue organization, enabling zero-shot retrieval of the tissue hierarchy. Pretrained protein representations can be adapted for downstream tasks: enhancing 3D structure-based representations for resolving immuno-oncological protein interactions, and investigating drugs' effects across cell types. Pinnacle outperforms state-of-the-art models in nominating therapeutic targets for rheumatoid arthritis and inflammatory bowel diseases, and pinpoints cell type contexts with higher predictive capability than context-free models. Pinnacle's ability to adjust its outputs based on the context in which it operates paves way for large-scale context-specific predictions in biology.
Collapse
Affiliation(s)
- Michelle M. Li
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Yepeng Huang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Marissa Sumathipala
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Man Qing Liang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Alberto Valdeolivas
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Ashwin N. Ananthakrishnan
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Katherine Liao
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, USA
| | - Daniel Marbach
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Marinka Zitnik
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, Allston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Data Science Initiative, Cambridge, MA, USA
| |
Collapse
|
3
|
Benoit JM, Breznik JA, Ang JC, Bhakta H, Huynh A, Cowbrough B, Baker B, Heessels L, Lodhi S, Yan E, Ewusie J, Nazy I, Bramson J, Miller MS, Bernatsky S, Larché MJ, Bowdish DME. Immunomodulatory drugs have divergent effects on humoral and cellular immune responses to SARS-CoV-2 vaccination in people living with rheumatoid arthritis. Sci Rep 2023; 13:22846. [PMID: 38129594 PMCID: PMC10739702 DOI: 10.1038/s41598-023-50263-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023] Open
Abstract
Understanding the efficacy of SARS-CoV-2 vaccination in people on immunosuppressive drugs, including those with rheumatoid arthritis (RA), is critical for their protection. Vaccine induced protection requires antibodies, CD4+ T cells, and CD8+ T cells, but it is unclear if these are equally affected by immunomodulatory drugs. Here, we determined how humoral and cellular SARS-CoV-2 vaccination responses differed between people with RA and controls, and which drug classes impacted these responses. Blood was collected from participants with RA on immunomodulatory drugs and controls after their second, third, and fourth SARS-CoV-2 vaccinations. Receptor binding domain (RBD)-specific antibodies were quantified by ELISA. Spike-specific memory T cells were quantitated using flow cytometry. Linear mixed models assessed the impact of age, sex, and immunomodulatory drug classes on SARS-CoV-2 vaccination responses. Compared to non-RA controls (n = 35), participants with RA on immunomodulatory drugs (n = 62) had lower anti-RBD IgG and spike-specific CD4+ T cell levels, but no deficits in spike-specific CD8+ T cells, following SARS-CoV-2 vaccination. Use of costimulation inhibitors was associated with lower humoral responses. JAK inhibitors were associated with fewer spike-specific CD4+ T cells. Participants with RA on immunomodulatory drugs mounted weaker responses to SARS-CoV-2 vaccination, with different drug classes impacting the cellular and humoral compartments.
Collapse
Affiliation(s)
- Jenna M Benoit
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, Hamilton, ON, Canada
- Firestone Institute of Respiratory Health, St. Joseph's Healthcare, Hamilton, ON, Canada
| | - Jessica A Breznik
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, Hamilton, ON, Canada
- McMaster Institute for Research on Aging, McMaster University, Hamilton, ON, Canada
| | - Jann C Ang
- Department of Biochemistry, McMaster University, Hamilton, ON, Canada
| | - Hina Bhakta
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Angela Huynh
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Braeden Cowbrough
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, Hamilton, ON, Canada
| | - Barbara Baker
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Lauren Heessels
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Sumiya Lodhi
- Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Elizabeth Yan
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Joycelyne Ewusie
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, ON, Canada
| | - Ishac Nazy
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Jonathan Bramson
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, Hamilton, ON, Canada
| | - Matthew S Miller
- McMaster Immunology Research Centre, Hamilton, ON, Canada
- Department of Biochemistry, McMaster University, Hamilton, ON, Canada
| | - Sasha Bernatsky
- Department of Medicine, McGill University, Montreal, QC, Canada
| | - Maggie J Larché
- Department of Medicine, McMaster University, Hamilton, ON, Canada.
- McMaster Immunology Research Centre, Hamilton, ON, Canada.
| | - Dawn M E Bowdish
- Department of Medicine, McMaster University, Hamilton, ON, Canada.
- McMaster Immunology Research Centre, Hamilton, ON, Canada.
- Firestone Institute of Respiratory Health, St. Joseph's Healthcare, Hamilton, ON, Canada.
| |
Collapse
|
4
|
Haroun E, Agrawal K, Leibovitch J, Kassab J, Zoghbi M, Dutta D, Lim SH. Chronic graft-versus-host disease in pediatric patients: Differences and challenges. Blood Rev 2023; 60:101054. [PMID: 36805299 DOI: 10.1016/j.blre.2023.101054] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023]
Abstract
Despite the use of high-resolution molecular techniques for tissue typing, chronic graft-versus-host disease (cGVHD) remains a major complication following allogeneic hematopoietic stem cell transplant. cGVHD adversely affects the life-expectancy and quality of life. The latter is particularly important and functionally relevant in pediatric patients who have a longer life-expectancy than adults. Current laboratory evidence suggests that there is not any difference in the pathophysiology of cGVHD between adults and pediatric patients. However, there are some clinical features and complications of the disease that are different in pediatric patients. There are also challenges in the development of new therapeutics for this group of patients. In this review, we will discuss the epidemiology, pathophysiology, clinical features and consequences of the disease, and highlight the differences between pediatric and adult patients. We will examine the current treatment options for pediatric patients with moderate to severe cGVHD and discuss the challenges facing therapeutic development for cGVHD in the pediatric population.
Collapse
Affiliation(s)
- Elio Haroun
- Division of Hematology and Oncology, State University of New York Upstate Medical University, Syracuse, NY, United States of America
| | - Kavita Agrawal
- Division of Hematology and Oncology, State University of New York Upstate Medical University, Syracuse, NY, United States of America
| | - Jennifer Leibovitch
- Division of Hematology and Oncology, State University of New York Upstate Medical University, Syracuse, NY, United States of America
| | - Joseph Kassab
- Department of Medicine, Saint-Joseph University of Beirut, Beirut, Lebanon
| | - Marianne Zoghbi
- Department of Medicine, Saint-Joseph University of Beirut, Beirut, Lebanon
| | - Dibyendu Dutta
- Division of Hematology and Oncology, State University of New York Upstate Medical University, Syracuse, NY, United States of America
| | - Seah H Lim
- Division of Hematology and Oncology, State University of New York Upstate Medical University, Syracuse, NY, United States of America,; Sanofi Oncology, Cambridge, MA, United States of America.
| |
Collapse
|
5
|
Algeri M, Becilli M, Locatelli F. Ruxolitinib as the first post-steroid treatment for acute and chronic graft-versus-host disease. Expert Rev Clin Immunol 2023; 19:1299-1313. [PMID: 37606511 DOI: 10.1080/1744666x.2023.2249230] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 08/14/2023] [Indexed: 08/23/2023]
Abstract
INTRODUCTION Acute and chronic graft-versus-host disease (GvHD) are potentially life-threatening complications occurring after allogeneic stem cell transplantation (allo-HSCT). Although steroids represent the first-line treatment for both conditions, in those patients who do not adequately benefit from steroid therapy, standardized treatment algorithms are lacking. In recent years, ruxolitinib has emerged as the most promising agent for the second-line therapy of steroid-refractory (SR)-GvHD. AREAS COVERED This review will summarize the biological properties and the mechanistic aspects that justify the therapeutic role of ruxolitinib in GvHD. In addition, current treatment options for SR-GvHD will be briefly discussed. Finally, results of the most relevant clinical trials on the use of ruxolitinib for SR-GvHD will be analyzed, with a particular focus on two phase-III randomized trials in which ruxolitinib demonstrated its superiority in comparison with the best available therapy. EXPERT OPINION Ruxolitinib has considerably improved the outcome of patients with SR-acute/chronic-GvHD and should be regarded as the standard-of-care option when corticosteroids fail or cannot be tapered. Nevertheless, a number of questions still remain unanswered and significant room for improvement exists. Additional observations derived from a longer follow-up will certainly increase our expertise in the management of this powerful therapy.
Collapse
Affiliation(s)
- Mattia Algeri
- Department of Haematology/Oncology and Cell and Gene Therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
- Department of Health Science, Magna Grecia University of Catanzaro, Catanzaro, Italy
| | - Marco Becilli
- Department of Haematology/Oncology and Cell and Gene Therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Franco Locatelli
- Department of Haematology/Oncology and Cell and Gene Therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
- Department of Life Sciences and Public Health, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
6
|
Holtan SG, Savid-Frontera C, Walton K, Eaton AA, Demorest C, Hoeschen A, Zhang L, Reid K, Kurian T, Sayegh Z, Julia E, Maakaron J, Bachanova V, Jurdi NE, MacMillan ML, Weisdorf DJ, Felices M, Miller JS, Blazar BR, Davila ML, Betts BC. Human Effectors of Acute and Chronic GVHD Overexpress CD83 and Predict Mortality. Clin Cancer Res 2023; 29:1114-1124. [PMID: 36622700 PMCID: PMC10011883 DOI: 10.1158/1078-0432.ccr-22-2837] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/31/2022] [Accepted: 01/05/2023] [Indexed: 01/10/2023]
Abstract
PURPOSE Acute and chronic GVHD remain major causes of transplant-related morbidity and mortality (TRM) after allogeneic hematopoietic cell transplantation (alloHCT). We have shown CD83 chimeric antigen receptor (CAR) T cells prevent GVHD and kill myeloid leukemia cell lines. In this pilot study, we investigate CD83 expression on GVHD effector cells, correlate these discoveries with clinical outcomes, and evaluate critical therapeutic implications for transplant recipients. EXPERIMENTAL DESIGN CD83 expression was evaluated among circulating CD4+ T cells, B-cell subsets, T follicular helper (Tfh) cells, and monocytes from patients with/without acute or chronic GVHD (n = 48 for each group), respectively. CD83 expression was correlated with survival, TRM, and relapse after alloHCT. Differential effects of GVHD therapies on CD83 expression was determined. RESULTS CD83 overexpression on CD4+ T cells correlates with reduced survival and increased TRM. Increased CD83+ B cells and Tfh cells, but not monocytes, are associated with poor posttransplant survival. CD83 CAR T eliminate autoreactive CD83+ B cells isolated from patients with chronic GVHD, without B-cell aplasia as observed with CD19 CAR T. We demonstrate robust CD83 antigen density on human acute myeloid leukemia (AML), and confirm potent antileukemic activity of CD83 CAR T in vivo, without observed myeloablation. CONCLUSIONS CD83 is a promising diagnostic marker of GVHD and warrants further investigation as a therapeutic target of both GVHD and AML relapse after alloHCT.
Collapse
Affiliation(s)
- Shernan G. Holtan
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Constanza Savid-Frontera
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kelly Walton
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Anne A. Eaton
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Connor Demorest
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Andrea Hoeschen
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Ling Zhang
- Department of Hematopathology and Laboratory Medicine, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kayla Reid
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Tony Kurian
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Zena Sayegh
- Department of Hematopathology and Laboratory Medicine, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Estefania Julia
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Joseph Maakaron
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Veronika Bachanova
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Najla El Jurdi
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Margaret L. MacMillan
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Daniel J. Weisdorf
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Martin Felices
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Jeffrey S. Miller
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Bruce R. Blazar
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Marco L. Davila
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Brian C. Betts
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
7
|
Yuan X, Jiang H, Fu D, Robida A, Rajanayake K, Yuan H, Wen B, Sun D, Watch BT, Chinnaswamy K, Stuckey JA, Paczesny S, Rech JC, Yang CY. Structure-Activity relationship of 1-(Furan-2ylmethyl)Pyrrolidine-Based Stimulation-2 (ST2) inhibitors for treating graft versus host disease. Bioorg Med Chem 2022; 71:116942. [PMID: 35930851 PMCID: PMC9451522 DOI: 10.1016/j.bmc.2022.116942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/12/2022] [Accepted: 07/16/2022] [Indexed: 11/20/2022]
Abstract
An elevated plasma level of soluble ST2 (sST2) is a risk biomarker for graft-versus-host disease (GVHD) and death in patients receiving hematopoietic cell transplantation (HCT). sST2 functions as a trap for IL-33 and amplifies the pro-inflammatory type 1 and 17 response while suppressing the tolerogenic type 2 and regulatory T cells activation during GVHD development. We previously identified small-molecule ST2 inhibitors particularly iST2-1 that reduces plasma sST2 levels and improved survival in two animal models. Here, we reported the structure-activity relationship of the furanylmethylpyrrolidine-based ST2 inhibitors based on iST2-1. Based on the biochemical AlphaLISA assay, we improved the activity of iST2-1 by 6-fold (∼6 μM in IC50 values) in the inhibition of ST2/IL-33 and confirmed the activities of the compounds in a cellular reporter assay. To determine the inhibition of the alloreactivity in vitro, we used the mixed lymphocyte reaction assay to demonstrate that our ST2 inhibitors decreased CD4+ and CD8+ T cells proliferation and increased Treg population. The data presented in this work are critical to the development of ST2 inhibitors in future.
Collapse
Affiliation(s)
- Xinrui Yuan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Hua Jiang
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Denggang Fu
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Aaron Robida
- Life Sciences Institute, Departments of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Krishani Rajanayake
- Rogel Cancer Center, Departments of Internal Medicine, University of Michigan, Ann Arbor, MI, United States; Pharmaceutical Sciences, College of Pharmacy, Departments of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Hebao Yuan
- Rogel Cancer Center, Departments of Internal Medicine, University of Michigan, Ann Arbor, MI, United States; Pharmaceutical Sciences, College of Pharmacy, Departments of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Bo Wen
- Rogel Cancer Center, Departments of Internal Medicine, University of Michigan, Ann Arbor, MI, United States; Pharmaceutical Sciences, College of Pharmacy, Departments of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Duxin Sun
- Rogel Cancer Center, Departments of Internal Medicine, University of Michigan, Ann Arbor, MI, United States; Pharmaceutical Sciences, College of Pharmacy, Departments of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Brennan T Watch
- Michigan Center for Therapeutic Innovation, Departments of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Krishnapriya Chinnaswamy
- Life Sciences Institute, Departments of Internal Medicine, University of Michigan, Ann Arbor, MI, United States; Rogel Cancer Center, Departments of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Jeanne A Stuckey
- Life Sciences Institute, Departments of Internal Medicine, University of Michigan, Ann Arbor, MI, United States; Rogel Cancer Center, Departments of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Sophie Paczesny
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Jason C Rech
- Michigan Center for Therapeutic Innovation, Departments of Internal Medicine, University of Michigan, Ann Arbor, MI, United States.
| | - Chao-Yie Yang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, United States.
| |
Collapse
|
8
|
JAK2 gene knockout inhibits corneal allograft rejection in mice by regulating dendritic cell-induced T cell immune tolerance. Cell Death Dis 2022; 8:289. [PMID: 35710633 PMCID: PMC9203759 DOI: 10.1038/s41420-022-01067-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 05/09/2022] [Accepted: 05/24/2022] [Indexed: 11/16/2022]
Abstract
Corneal allograft rejection can be seen in some patients after corneal transplantation. The present study intends to investigate whether JAK2 gene knockout affects corneal allograft rejection through regulation of dendritic cells (DCs)-induced T cell immune tolerance. In order to identify the target gene related to corneal allograft rejection, high-throughput mRNA sequencing and bioinformatics analysis were performed. JAK2 knockout mice were constructed and subjected to corneal allograft transplantation. The incidence of immune rejection was observed, the percentage of CD4+ T cells was detected, and the expression of Th1 cytokine interferon γ (IFN-γ) was determined. Flow cytometry and ELISA were performed to analyze the effects of JAK2 gene knockout on bone marrow-derived DCs (BMDCs). JAK2 was the target gene related to corneal allograft rejection. JAK2 gene knockout contributed to significantly prolonged survival time of corneal grafts in mice and inhibited corneal allograft rejection. The in vitro cell experiment further confirmed that JAK2 gene knockout contributed to the inactivation of CD4+ T cells and induced IFN-γ expression, accompanied by inhibition of DC immune function, development, maturation, and secretion of inflammatory cytokines. Collectively, JAK2 gene knockout inactivates CD4+ T cells to decrease IFN-γ expression, as well as inhibits DC development, maturation, and secretion of inflammatory cytokines, thereby reducing corneal allograft rejection.
Collapse
|
9
|
Walton K, Walker K, Riddle M, Koehn BH, Reff J, Sagatys EM, Linden MA, Pidala J, Kim J, Lee MC, Kiluk JV, Hui JYC, Yun SY, Xing Y, Stefanski H, Lawrence HR, Lawrence NJ, Tolar J, Anasetti C, Blazar BR, Sebti SM, Betts BC. Dual JAK2/Aurora kinase A inhibition prevents human skin graft rejection by allo-inactivation and ILC2-mediated tissue repair. Am J Transplant 2022; 22:717-730. [PMID: 34668635 PMCID: PMC8897228 DOI: 10.1111/ajt.16870] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 09/30/2021] [Accepted: 10/15/2021] [Indexed: 01/25/2023]
Abstract
Prevention of allograft rejection often requires lifelong immune suppression, risking broad impairment of host immunity. Nonselective inhibition of host T cell function increases recipient risk of opportunistic infections and secondary malignancies. Here we demonstrate that AJI-100, a dual inhibitor of JAK2 and Aurora kinase A, ameliorates skin graft rejection by human T cells and provides durable allo-inactivation. AJI-100 significantly reduces the frequency of skin-homing CLA+ donor T cells, limiting allograft invasion and tissue destruction by T effectors. AJI-100 also suppresses pathogenic Th1 and Th17 cells in the spleen yet spares beneficial regulatory T cells. We show dual JAK2/Aurora kinase A blockade enhances human type 2 innate lymphoid cell (ILC2) responses, which are capable of tissue repair. ILC2 differentiation mediated by GATA3 requires STAT5 phosphorylation (pSTAT5) but is opposed by STAT3. Further, we demonstrate that Aurora kinase A activation correlates with low pSTAT5 in ILC2s. Importantly, AJI-100 maintains pSTAT5 levels in ILC2s by blocking Aurora kinase A and reduces interference by STAT3. Therefore, combined JAK2/Aurora kinase A inhibition is an innovative strategy to merge immune suppression with tissue repair after transplantation.
Collapse
Affiliation(s)
- Kelly Walton
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Kirsti Walker
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Megan Riddle
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Brent H. Koehn
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Jordan Reff
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, USA
| | - Elizabeth M. Sagatys
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, USA,Department of Hematopathology and Laboratory Medicine, Moffitt Cancer Center, Tampa, FL, USA
| | - Michael A. Linden
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Joseph Pidala
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, USA,Department of Oncologic Sciences, Moffitt Cancer Center, Tampa, FL, USA,Department of Blood and Marrow Transplantation – Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Jongphil Kim
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
| | - Marie C Lee
- Department of Oncologic Sciences, Moffitt Cancer Center, Tampa, FL, USA,Department of Breast Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - John V. Kiluk
- Department of Oncologic Sciences, Moffitt Cancer Center, Tampa, FL, USA,Department of Breast Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | | | - Sang Y. Yun
- Department of Oncologic Sciences, Moffitt Cancer Center, Tampa, FL, USA,Department of Drug Discovery, Moffitt Cancer Center, Tampa, FL, USA
| | - Yan Xing
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Heather Stefanski
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Harshani R. Lawrence
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA,Department of Drug Discovery, Moffitt Cancer Center, Tampa, FL, USA
| | - Nicholas J. Lawrence
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA,Department of Drug Discovery, Moffitt Cancer Center, Tampa, FL, USA
| | - Jakub Tolar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Claudio Anasetti
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, USA,Department of Oncologic Sciences, Moffitt Cancer Center, Tampa, FL, USA,Department of Blood and Marrow Transplantation – Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Bruce R. Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Said M. Sebti
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia USA
| | - Brian C. Betts
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
10
|
Campe J, Ullrich E. T Helper Cell Lineage-Defining Transcription Factors: Potent Targets for Specific GVHD Therapy? Front Immunol 2022; 12:806529. [PMID: 35069590 PMCID: PMC8766661 DOI: 10.3389/fimmu.2021.806529] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
Allogenic hematopoietic stem cell transplantation (allo-HSCT) represents a potent and potentially curative treatment for many hematopoietic malignancies and hematologic disorders in adults and children. The donor-derived immunity, elicited by the stem cell transplant, can prevent disease relapse but is also responsible for the induction of graft-versus-host disease (GVHD). The pathophysiology of acute GVHD is not completely understood yet. In general, acute GVHD is driven by the inflammatory and cytotoxic effect of alloreactive donor T cells. Since several experimental approaches indicate that CD4 T cells play an important role in initiation and progression of acute GVHD, the contribution of the different CD4 T helper (Th) cell subtypes in the pathomechanism and regulation of the disease is a central point of current research. Th lineages derive from naïve CD4 T cell progenitors and lineage commitment is initiated by the surrounding cytokine milieu and subsequent changes in the transcription factor (TF) profile. Each T cell subtype has its own effector characteristics, immunologic function, and lineage specific cytokine profile, leading to the association with different immune responses and diseases. Acute GVHD is thought to be mainly driven by the Th1/Th17 axis, whereas Treg cells are attributed to attenuate GVHD effects. As the differentiation of each Th subset highly depends on the specific composition of activating and repressing TFs, these present a potent target to alter the Th cell landscape towards a GVHD-ameliorating direction, e.g. by inhibiting Th1 and Th17 differentiation. The finding, that targeting of Th1 and Th17 differentiation appears more effective for GVHD-prevention than a strategy to inhibit Th1 and Th17 cytokines supports this concept. In this review, we shed light on the current advances of potent TF inhibitors to alter Th cell differentiation and consecutively attenuate GVHD. We will focus especially on preclinical studies and outcomes of TF inhibition in murine GVHD models. Finally, we will point out the possible impact of a Th cell subset-specific immune modulation in context of GVHD.
Collapse
Affiliation(s)
- Julia Campe
- Experimental Immunology, Children's University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany.,Children's University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Evelyn Ullrich
- Experimental Immunology, Children's University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany.,Children's University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany.,Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Germany.,German Cancer Consortium (Deutsches Konsortium für Translationale Krebsforschung (DKTK)), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
| |
Collapse
|
11
|
Patel DA, Schroeder MA, Choi J, DiPersio JF. Mouse models of graft-versus-host disease. Methods Cell Biol 2022; 168:41-66. [DOI: 10.1016/bs.mcb.2021.12.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
12
|
Braun LM, Zeiser R. Kinase Inhibition as Treatment for Acute and Chronic Graft- Versus-Host Disease. Front Immunol 2021; 12:760199. [PMID: 34868001 PMCID: PMC8635802 DOI: 10.3389/fimmu.2021.760199] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/28/2021] [Indexed: 01/25/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HCT) is a potentially curative therapy for patients suffering from hematological malignancies via the donor immune system driven graft-versus-leukemia effect. However, the therapy is mainly limited by severe acute and chronic graft-versus-host disease (GvHD), both being life-threatening complications after allo-HCT. GvHD develops when donor T cells do not only recognize remaining tumor cells as foreign, but also the recipient’s tissue, leading to a severe inflammatory disease. Typical GvHD target organs include the skin, liver and intestinal tract. Currently all approved strategies for GvHD treatment are immunosuppressive therapies, with the first-line therapy being glucocorticoids. However, therapeutic options for glucocorticoid-refractory patients are still limited. Novel therapeutic approaches, which reduce GvHD severity while preserving GvL activity, are urgently needed. Targeting kinase activity with small molecule inhibitors has shown promising results in preclinical animal models and clinical trials. Well-studied kinase targets in GvHD include Rho-associated coiled-coil-containing kinase 2 (ROCK2), spleen tyrosine kinase (SYK), Bruton’s tyrosine kinase (BTK) and interleukin-2-inducible T-cell kinase (ITK) to control B- and T-cell activation in acute and chronic GvHD. Janus Kinase 1 (JAK1) and 2 (JAK2) are among the most intensively studied kinases in GvHD due to their importance in cytokine production and inflammatory cell activation and migration. Here, we discuss the role of kinase inhibition as novel treatment strategies for acute and chronic GvHD after allo-HCT.
Collapse
Affiliation(s)
- Lukas M Braun
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Center Freiburg (CCCF), University of Freiburg, Freiburg, Germany.,Centre for Biological Signalling Studies (BIOSS) and Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany
| |
Collapse
|
13
|
Jiang H, Fu D, Bidgoli A, Paczesny S. T Cell Subsets in Graft Versus Host Disease and Graft Versus Tumor. Front Immunol 2021; 12:761448. [PMID: 34675938 PMCID: PMC8525316 DOI: 10.3389/fimmu.2021.761448] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/13/2021] [Indexed: 01/04/2023] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is an essential therapeutic modality for patients with hematological malignancies and other blood disorders. Unfortunately, acute graft-versus-host disease (aGVHD) remains a major source of morbidity and mortality following allo-HCT, which limits its use in a broader spectrum of patients. Chronic graft-versus-host disease (cGVHD) also remains the most common long-term complication of allo-HCT, occurring in reportedly 30-70% of patients surviving more than 100 days. Chronic GVHD is also the leading cause of non-relapse mortality (NRM) occurring more than 2 years after HCT for malignant disease. Graft versus tumor (GVT) is a major component of the overall beneficial effects of allogeneic HCT in the treatment of hematological malignancies. Better understanding of GVHD pathogenesis is important to identify new therapeutic targets for GVHD prevention and therapy. Emerging data suggest opposing roles for different T cell subsets, e.g., IFN-γ producing CD4+ and CD8+ T cells (Th1 and Tc1), IL-4 producing T cells (Th2 and Tc2), IL-17 producing T cells (Th17 and Tc17), IL-9 producing T cells (Th9 and Tc9), IL-22 producing T cells (Th22), T follicular helper cells (Tfh), regulatory T-cells (Treg) and tissue resident memory T cells (Trm) in GVHD and GVT etiology. In this review, we first summarize the general description of the cytokine signals that promote the differentiation of T cell subsets and the roles of these T cell subsets in the pathogenesis of GVHD. Next, we extensively explore preclinical findings of T cell subsets in both GVHD/GVT animal models and humans. Finally, we address recent findings about the roles of T-cell subsets in clinical GVHD and current strategies to modulate T-cell differentiation for treating and preventing GVHD in patients. Further exploring and outlining the immune biology of T-cell differentiation in GVHD that will provide more therapeutic options for maintaining success of allo-HCT.
Collapse
Affiliation(s)
| | | | | | - Sophie Paczesny
- Department of Microbiology and Immunology and Pediatrics, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
14
|
Peritransplant Ruxolitinib Administration is Safe and Effective in Patients with Myelofibrosis: a Pilot Open-Label Study. Blood Adv 2021; 6:1444-1453. [PMID: 34581764 PMCID: PMC8905711 DOI: 10.1182/bloodadvances.2021005035] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/14/2021] [Indexed: 11/20/2022] Open
Abstract
Peritransplantation ruxolitinib was safe and well tolerated in patients with myelofibrosis, with MTD determined as 10 mg twice daily.
We report results of our prospective pilot trial evaluating safety/feasibility of peritransplantation ruxolitinib for myelofibrosis treatment. Primary objectives were to determine safety and maximum tolerated dose (MTD) of ruxolitinib. Ruxolitinib was administered at 2 dose levels (DLs) of 5 and 10 mg twice daily, with fludarabine/melphalan conditioning regimen and tacrolimus/sirolimus graft-versus-host disease (GVHD) prophylaxis. We enrolled 6 and 12 patients at DL1 and DL2, respectively. Median age at transplantation was 65 years (range, 25-73). Per Dynamic International Prognostic Scoring System, 4 patients were high and 14 intermediate risk. Peripheral blood stem cells were graft source from matched sibling (n = 5) or unrelated (n = 13) donor. At each DL, 1 patient developed dose-limiting toxicities (DLTs): grade 3 cardiac and gastrointestinal with grade 4 pulmonary DLTs in DL1, and grade 3 kidney injury in DL2. All patients achieved engraftment. Grade 2 to 4 and 3 to 4 acute GVHD cumulative incidence was 17% (95% confidence interval [CI], 6-47) and 11% (95% CI, 3-41), respectively. Cumulative incidence of 1-year chronic GVHD was 42% (95% CI, 24-74). With 22.6-month (range, 6.2-25.8) median follow-up in surviving patients, 1-year overall and progression-free survival were 77% (95% CI, 50-91) and 71% (95% CI, 44-87), respectively. Causes of death (n = 4) were cardiac arrest, GVHD, respiratory failure, and refractory GVHD of liver. Our results show peritransplantation ruxolitinib is safe and well tolerated at MTD of 10 mg twice daily and associated with dose-dependent pharmacokinetic and cytokine profile. Early efficacy data are highly promising in high-risk older patients with myelofibrosis. This trial was registered at www.clinicaltrials.gov as #NCT02917096.
Collapse
|
15
|
Tzeng HT, Chyuan IT, Lai JH. Targeting the JAK-STAT pathway in autoimmune diseases and cancers: A focus on molecular mechanisms and therapeutic potential. Biochem Pharmacol 2021; 193:114760. [PMID: 34492272 DOI: 10.1016/j.bcp.2021.114760] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 01/01/2023]
Abstract
The Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathway is characterized by diverse immune regulatory systems involving cell proliferation, survival, and inflammation and immune tolerance. Aberrant JAK/STAT transduction activates proinflammatory cytokine signaling that jeopardize the immune balance and thus contributes to the development of autoimmune diseases and cancer progression. The success of several small-molecule JAK inhibitors in the treatment of rheumatologic diseases demonstrates that targeting the JAK/STAT pathway is efficient in suppressing inflammation and sheds light on their therapeutic potential in several autoimmune diseases and cancers. In this review, we discuss the signal transduction and molecular mechanism involving immune function through the JAK-STAT pathway, outline the role of this pathway in autoimmunity and oncoimmunology, and explain the preclinical and clinical trial evidence for the therapeutic potential of targeting the JAK-STAT signaling pathway. Issues regarding the safety and clinical efficacy of JAK inhibitors are reviewed. Ongoing studies are addressed with a focus on emerging indications for JAK inhibition and explanations of the novel mechanisms of JAK-STAT signaling blockade.
Collapse
Affiliation(s)
- Hong-Tai Tzeng
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - I-Tsu Chyuan
- Department of Internal Medicine, Cathay General Hospital, Taipei, Taiwan; Department of Medical Research, Cathay General Hospital, Taipei, Taiwan; School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan.
| | - Jenn-Haung Lai
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Tao-Yuan, Taiwan; Graduate Institute of Medical Science, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
16
|
Cyriac S, Prem S, Salas MQ, Chen S, Al-Shaibani Z, Lam W, Law A, Gupta V, Michelis FV, Kim DDH, Lipton J, Kumar R, Mattsson J, Viswabandya A. Effect of pre-transplant JAK1/2 inhibitors and CD34 dose on transplant outcomes in myelofibrosis. Eur J Haematol 2021; 107:517-528. [PMID: 34260760 DOI: 10.1111/ejh.13689] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/11/2021] [Accepted: 07/13/2021] [Indexed: 11/30/2022]
Abstract
Allogeneic hematopoeitic cell transplantation (allo-HCT) is the only curative treatment for myelofibrosis (MF). We evaluate the impact of various factors on survival outcomes post-transplant in MF. Data of 89 consecutive MF patients (primary 47%) who underwent allo-HCT between 2005 and 2018 was evaluated. Fifty-four percent patients had received JAK1/2 inhibitors (JAKi) pre-HCT. The median CD34 count was 7.1x106 cells/kg. Graft failure was seen in 10% of the patients. Grade 3-4 acute GVHD (aGVHD) and moderate/severe chronic graft versus host disease (cGVHD) occurred in 24% and 40% patients, respectively. Two-year overall survival (OS) and relapse free survival (RFS) were 51% and 43%, respectively. Cumulative incidence of relapse (CIR) and non-relapse mortality (NRM) at 2 years were 11% and 46%, respectively. Higher CD34 cell dose (≤5 × 106 cells/kg vs 5-9 or ≥9 × 106 cells/kg) and lower pre-HCT ferritin (</=1000 ng/ml) were associated with better OS, RFS and lower NRM. Grade 3-4 aGVHD was associated with higher NRM. Use of pre-transplant JAKi was associated with lower incidence of grade 3-4 aGVHD. In summary, higher CD34 cell dose is associated with better allo-HCT outcomes in MF and pre-HCT JAKi use is associated with reduced risk of severe aGVHD. These two modifiable parameters should be considered during allo-HCT for MF.
Collapse
Affiliation(s)
- Sunu Cyriac
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Shruti Prem
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Maria Queralt Salas
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Shiyi Chen
- Department of Biostatistics, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Zeyad Al-Shaibani
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Wilson Lam
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Arjun Law
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Vikas Gupta
- Leukemia Program, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Fotios V Michelis
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Dennis Dong Hwan Kim
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Jeffrey Lipton
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Rajat Kumar
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Jonas Mattsson
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Auro Viswabandya
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
17
|
Huarte E, Peel M, Juvekar A, Dubé P, Sarah S, Stephens L, Stewart B, Long B, Czerniak P, Oliver J, Smith P. Ruxolitinib, a JAK1/JAK2 selective inhibitor, ameliorates acute and chronic steroid-refractory GvHD mouse models. Immunotherapy 2021; 13:977-987. [PMID: 34184542 DOI: 10.2217/imt-2021-0013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Aim: Graft-versus-host disease (GvHD) is a major complication arising in patients undergoing allogenic hematopoietic stem cell transplantation. Material & methods: We tested ruxolitinib (a selective JAK1/2 inhibitor) efficacy in three different preclinical models of GvHD. Results: Ruxolitinib, at doses that mimic clinically achievable human JAK/signal transducers and activators of transcription target inhibition, significantly reduced alloreactive T-cell activation and infiltration in the lung and skin, leading to improved outcomes in two experimental models of steroid-refractory acute and chronic GvHD. Additionally, we describe a novel humanized GvHD model in which immunodeficient NOG animals are engineered to produce human IL-15 to facilitate enhanced T- and NK cell engraftment, leading to severe GvHD. Conclusion: Ruxolitinib treatment ameliorated disease symptoms resulting from targeted immune modulation via JAK/signal transducers and activators of transcription signaling inhibition.
Collapse
Affiliation(s)
- Eduardo Huarte
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19803, USA
| | - Michael Peel
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19803, USA
| | - Ashish Juvekar
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19803, USA
| | - Philip Dubé
- Taconic Biosciences, 1 Discovery Drive, Rensselaer, NY 12144, USA
| | - Sarala Sarah
- Taconic Biosciences, 1 Discovery Drive, Rensselaer, NY 12144, USA
| | - Lynn Stephens
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19803, USA
| | - Becky Stewart
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19803, USA
| | - Brian Long
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19803, USA
| | - Philip Czerniak
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19803, USA
| | - Julian Oliver
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19803, USA
| | - Paul Smith
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19803, USA
| |
Collapse
|
18
|
Untwining Anti-Tumor and Immunosuppressive Effects of JAK Inhibitors-A Strategy for Hematological Malignancies? Cancers (Basel) 2021; 13:cancers13112611. [PMID: 34073410 PMCID: PMC8197909 DOI: 10.3390/cancers13112611] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/18/2021] [Accepted: 05/22/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway is aberrantly activated in many malignancies. Inhibition of this pathway via JAK inhibitors (JAKinibs) is therefore an attractive therapeutic strategy underlined by Ruxolitinib (JAK1/2 inhibitor) being approved for the treatment of myeloproliferative neoplasms. As a consequence of the crucial role of the JAK-STAT pathway in the regulation of immune responses, inhibition of JAKs suppresses the immune system. This review article provides a thorough overview of the current knowledge on JAKinibs’ effects on immune cells in the context of hematological malignancies. We also discuss the potential use of JAKinibs for the treatment of diseases in which lymphocytes are the source of the malignancy. Abstract The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway propagates signals from a variety of cytokines, contributing to cellular responses in health and disease. Gain of function mutations in JAKs or STATs are associated with malignancies, with JAK2V617F being the main driver mutation in myeloproliferative neoplasms (MPN). Therefore, inhibition of this pathway is an attractive therapeutic strategy for different types of cancer. Numerous JAK inhibitors (JAKinibs) have entered clinical trials, including the JAK1/2 inhibitor Ruxolitinib approved for the treatment of MPN. Importantly, loss of function mutations in JAK-STAT members are a cause of immune suppression or deficiencies. MPN patients undergoing Ruxolitinib treatment are more susceptible to infections and secondary malignancies. This highlights the suppressive effects of JAKinibs on immune responses, which renders them successful in the treatment of autoimmune diseases but potentially detrimental for cancer patients. Here, we review the current knowledge on the effects of JAKinibs on immune cells in the context of hematological malignancies. Furthermore, we discuss the potential use of JAKinibs for the treatment of diseases in which lymphocytes are the source of malignancies. In summary, this review underlines the necessity of a robust immune profiling to provide the best benefit for JAKinib-treated patients.
Collapse
|
19
|
Ruxolitinib for the treatment of chronic GVHD and overlap syndrome in children and young adults. Transplantation 2021; 106:412-419. [PMID: 33795598 DOI: 10.1097/tp.0000000000003768] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Ruxolitinib, a JAK1/2 inhibitor, is used to treat chronic graft-versus-host-disease (cGVHD) in adult allogeneic hematopoietic stem cell transplant patients, but experience in children is limited, perhaps due to lack of pediatric dosing information. In this report we describe our pediatric and young adult dosing strategy experience in cGVHD. METHODS Ruxolitinib was administered orally at 5mg twice daily for children ≥25kg or 2.5mg twice daily if <25kg. The dose was halved with concurrent azole administration and increased to a maximum of 10mg twice daily if tolerated. Responses were evaluated using the 2014 NIH consensus criteria. Phosphorylation of lymphocyte STAT5 following dosing, a surrogate of JAK inhibition, was evaluated by flow cytometry. RESULTS Twenty patients with a median age 14.6 years (range 5-26 years) received ruxolitinib for severe (n=9) and moderate (n=11) cGVHD. Median steroid dose was 0.5mg/kg/day (range 0.08-1.5mg/kg/day) at ruxolitinib initiation. Two patients with moderate cGVHD achieved a complete response (CR), while 12 patients achieved a partial response (PR) at a median of 48 days (range 17-98 days) from first ruxolitinib dose, for an overall response rate of 70%. Eleven patients are maintaining their PRs. pSTAT5 on lymphocytes was absent or decreased (0-6% events) in 5 evaluated patients, suggesting adequate inhibition. Three patients discontinued ruxolitinib due to neutropenia, thrombocytopenia, or elevated alanine aminotransferase. Four patients developed bacterial infections, and three experienced symptomatic viral infections. Two patients died from complications related to progressive severe cGVHD. CONCLUSION Ruxolitinib using our dosing strategy demonstrates promise for treating cGVHD in children.Supplemental Visual Abstract; http://links.lww.com/TP/C202.
Collapse
|
20
|
Pidala J, Walton K, Elmariah H, Kim J, Mishra A, Bejanyan N, Nishihori T, Khimani F, Perez L, Faramand RG, Davila ML, Nieder ML, Sagatys EM, Holtan SG, Lawrence NJ, Lawrence HR, Blazar BR, Anasetti C, Sebti SM, Betts BC. Pacritinib Combined with Sirolimus and Low-Dose Tacrolimus for GVHD Prevention after Allogeneic Hematopoietic Cell Transplantation: Preclinical and Phase I Trial Results. Clin Cancer Res 2021; 27:2712-2722. [PMID: 33753457 DOI: 10.1158/1078-0432.ccr-20-4725] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/22/2021] [Accepted: 03/10/2021] [Indexed: 01/17/2023]
Abstract
PURPOSE In this first-in-human, phase I, GVHD prevention trial (NCT02891603), we combine pacritinib (PAC), a JAK2 inhibitor, with sirolimus to concurrently reduce T-cell costimulation via mTOR and IL6 activity. We evaluate the safety of pacritinib when administered with sirolimus plus low-dose tacrolimus (PAC/SIR/TAC) after allogeneic hematopoietic cell transplantation. PATIENTS AND METHODS The preclinical efficacy and immune modulation of PAC/SIR were investigated in xenogeneic GVHD. Our phase I trial followed a 3+3 dose-escalation design, including dose level 1 (pacritinib 100 mg daily), level 2 (pacritinib 100 mg twice daily), and level 3 (pacritinib 200 mg twice daily). The primary endpoint was to identify the lowest biologically active and safe dose of pacritinib with SIR/TAC (n = 12). Acute GVHD was scored through day +100. Allografts included 8/8 HLA-matched related or unrelated donor peripheral blood stem cells. RESULTS In mice, we show that dual JAK2/mTOR inhibition significantly reduces xenogeneic GVHD and increases peripheral regulatory T cell (Treg) potency as well as Treg induction from conventional CD4+ T cells. Pacritinib 100 mg twice a day was identified as the minimum biologically active and safe dose for further study. JAK2/mTOR inhibition suppresses pathogenic Th1 and Th17 cells, spares Tregs and antileukemia effector cells, and exhibits preliminary activity in preventing GVHD. PAC/SIR/TAC preserves donor cytomegalovirus (CMV) immunity and permits timely engraftment without cytopenias. CONCLUSIONS We demonstrate that PAC/SIR/TAC is safe and preliminarily limits acute GVHD, preserves donor CMV immunity, and permits timely engraftment. The efficacy of PAC/SIR/TAC will be tested in our ongoing phase II GVHD prevention trial.
Collapse
Affiliation(s)
- Joseph Pidala
- Department of Blood and Marrow Transplantation - Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida.,Department of Immunology, Moffitt Cancer Center, Tampa, Florida.,Department of Oncologic Sciences, University of South Florida, Tampa, Florida
| | - Kelly Walton
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Hany Elmariah
- Department of Blood and Marrow Transplantation - Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida.,Department of Oncologic Sciences, University of South Florida, Tampa, Florida
| | - Jongphil Kim
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida
| | - Asmita Mishra
- Department of Blood and Marrow Transplantation - Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida.,Department of Oncologic Sciences, University of South Florida, Tampa, Florida
| | - Nelli Bejanyan
- Department of Blood and Marrow Transplantation - Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida.,Department of Oncologic Sciences, University of South Florida, Tampa, Florida
| | - Taiga Nishihori
- Department of Blood and Marrow Transplantation - Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida.,Department of Oncologic Sciences, University of South Florida, Tampa, Florida
| | - Farhad Khimani
- Department of Blood and Marrow Transplantation - Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida.,Department of Oncologic Sciences, University of South Florida, Tampa, Florida
| | - Lia Perez
- Department of Blood and Marrow Transplantation - Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida.,Department of Oncologic Sciences, University of South Florida, Tampa, Florida
| | - Rawan G Faramand
- Department of Blood and Marrow Transplantation - Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida.,Department of Oncologic Sciences, University of South Florida, Tampa, Florida
| | - Marco L Davila
- Department of Blood and Marrow Transplantation - Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida.,Department of Immunology, Moffitt Cancer Center, Tampa, Florida.,Department of Oncologic Sciences, University of South Florida, Tampa, Florida
| | - Michael L Nieder
- Department of Blood and Marrow Transplantation - Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida.,Department of Oncologic Sciences, University of South Florida, Tampa, Florida
| | - Elizabeth M Sagatys
- Department of Hematopathology and Laboratory Medicine, Moffitt Cancer Center, Tampa, Florida
| | - Shernan G Holtan
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | | | | | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Claudio Anasetti
- Department of Blood and Marrow Transplantation - Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida.,Department of Immunology, Moffitt Cancer Center, Tampa, Florida.,Department of Oncologic Sciences, University of South Florida, Tampa, Florida
| | - Said M Sebti
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - Brian C Betts
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
21
|
Hammoudeh SM, Venkatachalam T, Ansari AW, Bendardaf R, Hamid Q, Rahmani M, Hamoudi R. Systems Immunology Analysis Reveals an Immunomodulatory Effect of Snail-p53 Binding on Neutrophil- and T Cell-Mediated Immunity in KRAS Mutant Non-Small Cell Lung Cancer. Front Immunol 2020; 11:569671. [PMID: 33381110 PMCID: PMC7768232 DOI: 10.3389/fimmu.2020.569671] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 11/11/2020] [Indexed: 11/13/2022] Open
Abstract
Immunomodulation and chronic inflammation are important mechanisms utilized by cancer cells to evade the immune defense and promote tumor progression. Therefore, various efforts were focused on the development of approaches to reprogram the immune response to increase the immune detection of cancer cells and enhance patient response to various types of therapy. A number of regulatory proteins were investigated and proposed as potential targets for immunomodulatory therapeutic approaches including p53 and Snail. In this study, we investigated the immunomodulatory effect of disrupting Snail-p53 binding induced by the oncogenic KRAS to suppress p53 signaling. We analyzed the transcriptomic profile mediated by Snail-p53 binding inhibitor GN25 in non-small cell lung cancer cells (A549) using Next generation whole RNA-sequencing. Notably, we observed a significant enrichment in transcripts involved in immune response pathways especially those contributing to neutrophil (IL8) and T-cell mediated immunity (BCL6, and CD81). Moreover, transcripts associated with NF-κB signaling were also enriched which may play an important role in the immunomodulatory effect of Snail-p53 binding. Further analysis revealed that the immune expression signature of GN25 overlaps with the signature of other therapeutic compounds known to exhibit immunomodulatory effects validating the immunomodulatory potential of targeting Snail-p53 binding. The effects of GN25 on the immune response pathways suggest that targeting Snail-p53 binding might be a potentially effective therapeutic strategy.
Collapse
Affiliation(s)
- Sarah Musa Hammoudeh
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Thenmozhi Venkatachalam
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Abdul Wahid Ansari
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Riyad Bendardaf
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Oncology Unit, University Hospital Sharjah, Sharjah, United Arab Emirates
| | - Qutayba Hamid
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
| | - Mohamed Rahmani
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Rifat Hamoudi
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,Division of Surgery and Interventional Science, University College London, London, United Kingdom
| |
Collapse
|
22
|
Vitamin D3 receptor polymorphisms regulate T cells and T cell-dependent inflammatory diseases. Proc Natl Acad Sci U S A 2020; 117:24986-24997. [PMID: 32958661 DOI: 10.1073/pnas.2001966117] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
It has proven difficult to identify the underlying genes in complex autoimmune diseases. Here, we use forward genetics to identify polymorphisms in the vitamin D receptor gene (Vdr) promoter, controlling Vdr expression and T cell activation. We isolated these polymorphisms in a congenic mouse line, allowing us to study the immunomodulatory properties of VDR in a physiological context. Congenic mice overexpressed VDR selectively in T cells, and thus did not suffer from calcemic effects. VDR overexpression resulted in an enhanced antigen-specific T cell response and more severe autoimmune phenotypes. In contrast, vitamin D3-deficiency inhibited T cell responses and protected mice from developing autoimmune arthritis. Our observations are likely translatable to humans, as Vdr is overexpressed in rheumatic joints. Genetic control of VDR availability codetermines the proinflammatory behavior of T cells, suggesting that increased presence of VDR at the site of inflammation might limit the antiinflammatory properties of its ligand.
Collapse
|
23
|
Shrestha B, Walton K, Reff J, Sagatys EM, Tu N, Boucher J, Li G, Ghafoor T, Felices M, Miller JS, Pidala J, Blazar BR, Anasetti C, Betts BC, Davila ML. Human CD83-targeted chimeric antigen receptor T cells prevent and treat graft-versus-host disease. J Clin Invest 2020; 130:4652-4662. [PMID: 32437331 PMCID: PMC7456225 DOI: 10.1172/jci135754] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 05/15/2020] [Indexed: 12/17/2022] Open
Abstract
Graft-versus-host disease (GVHD) remains an important cause of morbidity and mortality after allogeneic hematopoietic cell transplantation (allo-HCT). For decades, GVHD prophylaxis has included calcineurin inhibitors, despite their incomplete efficacy and impairment of graft-versus-leukemia (GVL). Distinct from pharmacologic immune suppression, we have developed what we believe is a novel, human CD83-targeted chimeric antigen receptor (CAR) T cell for GVHD prevention. CD83 is expressed on allo-activated conventional CD4+ T cells (Tconvs) and proinflammatory dendritic cells (DCs), which are both implicated in GVHD pathogenesis. Human CD83 CAR T cells eradicate pathogenic CD83+ target cells, substantially increase the ratio of regulatory T cells (Tregs) to allo-activated Tconvs, and provide durable prevention of xenogeneic GVHD. CD83 CAR T cells are also capable of treating xenogeneic GVHD. We show that human acute myeloid leukemia (AML) expresses CD83 and that myeloid leukemia cell lines are readily killed by CD83 CAR T cells. Human CD83 CAR T cells are a promising cell-based approach to preventing 2 critical complications of allo-HCT - GVHD and relapse. Thus, the use of human CD83 CAR T cells for GVHD prevention and treatment, as well as for targeting CD83+ AML, warrants clinical investigation.
Collapse
Affiliation(s)
- Bishwas Shrestha
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Kelly Walton
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jordan Reff
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Elizabeth M. Sagatys
- Department of Hematopathology and Laboratory Medicine, Moffitt Cancer Center, Tampa, Florida, USA
- Department of Oncologic Sciences, University of South Florida, Tampa, Florida, USA
| | - Nhan Tu
- Department of Oncologic Sciences, University of South Florida, Tampa, Florida, USA
| | - Justin Boucher
- Department of Oncologic Sciences, University of South Florida, Tampa, Florida, USA
| | - Gongbo Li
- Department of Oncologic Sciences, University of South Florida, Tampa, Florida, USA
| | - Tayyebb Ghafoor
- Department of Oncologic Sciences, University of South Florida, Tampa, Florida, USA
| | - Martin Felices
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jeffrey S. Miller
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Joseph Pidala
- Department of Oncologic Sciences, University of South Florida, Tampa, Florida, USA
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida, USA
| | - Bruce R. Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Claudio Anasetti
- Department of Oncologic Sciences, University of South Florida, Tampa, Florida, USA
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida, USA
| | - Brian C. Betts
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Marco L. Davila
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida, USA
- Department of Oncologic Sciences, University of South Florida, Tampa, Florida, USA
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida, USA
| |
Collapse
|
24
|
Betts BC, Young JW. Less Can Be More When Targeting Interleukin-6-Mediated Cytokine Release Syndrome in Coronavirus Disease 2019. Crit Care Explor 2020; 2:e0138. [PMID: 32696001 PMCID: PMC7314334 DOI: 10.1097/cce.0000000000000138] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Coronavirus disease 2019 pandemic caused by severe acute respiratory syndrome-coronavirus-2 is a worldwide public health emergency that will have a lasting generational impact in terms of mortality and economic devastation. Social distancing to prevent viral transmission and supportive care of infected patients are the main interventions now available. This global health crisis therefore merits innovative therapies. Cytokine release syndrome mediated by interleukin-6 is a critical driver of coronavirus disease 2019 mortality. Herein, we review and discuss key immunologic effects of direct interleukin-6 blockade, downstream nonselective Janus kinase inhibition, and selective Janus kinase 2 suppression to treat coronavirus disease 2019-related cytokine release syndrome. We provide evidence that selective targeting of interleukin-6 or Janus kinase 2 is well informed by existing data. This contrasts with broad, nonselective blockade of Janus kinase-mediated signaling, which would inhibit both deleterious and beneficial cytokines, as well as critical host antiviral immunity.
Collapse
Affiliation(s)
- Brian C Betts
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - James W Young
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
- The Rockefeller University, New York, NY
| |
Collapse
|
25
|
Ali H, Salhotra A, Modi B, Nakamura R. Ruxolitinib for the treatment of graft-versus-host disease. Expert Rev Clin Immunol 2020; 16:347-359. [DOI: 10.1080/1744666x.2020.1740592] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Haris Ali
- Department of Hematology/Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Amandeep Salhotra
- Department of Hematology/Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Badri Modi
- Department of Surgery, Division of Dermatology, City of Hope National Medical Center, Duarte, CA, USA
| | - Ryotaro Nakamura
- Department of Hematology/Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
26
|
The primacy of gastrointestinal tract antigen-presenting cells in lethal graft-versus-host disease. Blood 2020; 134:2139-2148. [PMID: 31697827 DOI: 10.1182/blood.2019000823] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/24/2019] [Indexed: 12/26/2022] Open
Abstract
Allogeneic stem cell transplantation is a cornerstone of curative therapy for high-risk and/or advanced hematological malignancies but remains limited by graft-versus-host disease (GVHD). GVHD is initiated by the interaction between recipient antigen-presenting cells (APCs) and donor T cells, culminating in T-cell differentiation along pathogenic type-1 and type-17 paradigms at the expense of tolerogenic regulatory T-cell patterns. Type-1 and type-17 T cells secrete cytokines (eg, granulocyte-macrophage colony-stimulating factor and interferon-γ) critical to the cytokine storm that amplifies expansion of donor APCs and their alloantigen presentation. It has become increasingly clear that pathogenic donor T-cell differentiation is initiated by both professional recipient APCs (eg, dendritic cells [DCs]) and nonprofessional APCs (eg, epithelial and mesenchymal cells), particularly within the gastrointestinal (GI) tract. In the immediate peritransplantation period, these APCs are profoundly modified by pathogen-associated molecular pattern (PAMP)/damage-associated molecular pattern (DAMP) signals derived from conditioning and intestinal microbiota. Subsequently, donor DCs in the GI tract are activated by DAMP/PAMP signals in the colon that gain access to the lamina propria once the mucosal barrier mucosa is compromised by GVHD. This results in donor DC expansion and alloantigen presentation in the colon and subsequent migration into the mesenteric lymph nodes. Here, new donor T cells are primed, expanded, differentiated, and imprinted with gut-homing integrins permissive of migration into the damaged GI tract, resulting in the lethal feed-forward cascade of GVHD. These new insights into our understanding of the cellular and molecular factors initiating GVHD, both spatially and temporally, give rise to a number of logical therapeutic targets, focusing on the inhibition of APC function in the GI tract.
Collapse
|
27
|
Mannina D, Kröger N. Janus Kinase Inhibition for Graft-Versus-Host Disease: Current Status and Future Prospects. Drugs 2020; 79:1499-1509. [PMID: 31359326 DOI: 10.1007/s40265-019-01174-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (Allo-HSCT) is a curative treatment for many hematological malignant and non-malignant diseases. A major complication of the procedure is the donor T-cell-mediated graft-versus-host disease (GvHD). GvHD accounts for about 10% of early mortality after transplantation. GVHD is also the major cause of morbidity and disability in the late follow-up phase of transplanted patients, mainly because of the low response to first-line steroids, and the lack of efficient second-line standard treatments. The increasing knowledge regarding GVHD pathogenesis provides new pharmacological targets, potentially exploitable in clinical practice, in order to prevent and treat this complication. This review provides a description of GVHD pathogenesis, with a focus on the central role of the Janus kinase-related mechanisms. The first inflammatory innate-immunity response is triggered by a JAK/STAT dependent pathway, and JAK inhibition impairs antigen-presenting cell differentiation and activation and downregulates the expression of signals for T-cell triggering. The chronic evolution of alloreactivity, characterized by the long-term maintenance of inflammation and fibrosis, is also dependent on JAK/STAT activation. Based on preclinical data, we reviewed the rationale behind the clinical use of JAK-inhibitors in GVHD, presenting available results of clinical trials and reports, and looked at future implementation of this new promising treatment approach.
Collapse
Affiliation(s)
- Daniele Mannina
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
- Department of Hematology, Vita-Salute San Raffaele University Milano, Milan, Italy
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| |
Collapse
|
28
|
Zhao Y, Shi J, Luo Y, Gao F, Tan Y, Lai X, Yu J, Wei G, Huang H. Calcineurin Inhibitors Replacement by Ruxolitinib as Graft-versus-Host Disease Prophylaxis for Patients after Allogeneic Stem Cell Transplantation. Biol Blood Marrow Transplant 2020; 26:e128-e133. [PMID: 31982545 DOI: 10.1016/j.bbmt.2020.01.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/06/2020] [Accepted: 01/16/2020] [Indexed: 10/25/2022]
Abstract
Graft-versus-host disease (GVHD) is a common complication of allogeneic stem cell transplantation (allo-SCT) that carries a high mortality. Although calcineurin inhibitors (CNIs) have been widely used in GVHD prophylaxis, the incidence of acute GVHD (aGVHD) remains at roughly 30% to 50%. Moreover, some allo-SCT recipients cannot tolerate CNI. Thus, improved GVHD prevention methods are needed. Our study aimed to determine the prophylactic value of ruxolitinib for GVHD in CNI-intolerant patients after allo-SCT. Between September 2017 and March 2019, 10 patients with hematopoietic malignancies after allo-SCT who were intolerant to CNI at our center were enrolled in this study. The regimens were based on a myeloablative busulfan and cyclophosphamide regimen. Antithymocyte globulin was administered to patients with an HLA-haploidentical related donor (HRD) at a dosage of 6 mg/kg. All received ruxolitinib to replace CNI as GVHD prophylaxis. Ruxolitinib was initiated at 5 to 10 mg twice daily until 2 to 3 months post-transplantation and then tapered gradually, and in the absence of GVHD, discontinued by day +180. Eight patients had acute leukemia, 1 patient had myeloproliferative neoplasm, and 1 patient had natural killer T cell (NK/T) lymphoma. The donor type was a matched sibling donor in 3 patients and an HLA-haploidentical related donor (HRD) in 7 patients. All patients received CNI plus short-course of methotrexate as GVHD prophylaxis, but showed intolerance to CNI within 45 days post-transplantation. After ruxolitinib replacement, only 1 patient (10%) developed grade II skin aGVHD within 100 days, and only 1 patient developed severe aGVHD after 100 days. Two patients developed moderate/severe chronic GVHD (cGVHD) after tapering or stopping ruxolitinib, resulting in a 1-year cumulative incidence of moderate/severe cGVHD of 21.4%. Cytomegalovirus (CMV) reactivation occurred in 4 patients (40%), and Epstein-Barr virus (EBV) reactivation occurred in 3 patients (30%). None of the patients developed CMV disease or EBV post-transplantation lymphoproliferative disorder. After a median follow-up of 11 months (range, 2 to 15.5 months), 2 patients (20%) relapsed and 7 (70%) were alive, of whom 6 (60%) were negative for minimal residual disease and 4 were off immunosuppressant therapy. The prophylactic application of ruxolitinib for CNI-intolerant patients after allo-SCT appears to be safe and effective in preventing GVHD, but this awaits further study in larger cohorts.
Collapse
Affiliation(s)
- Yanmin Zhao
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jimin Shi
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yi Luo
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Fei Gao
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yamin Tan
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaoyu Lai
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jian Yu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Guoqing Wei
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
29
|
Chen JY, Lei SY, Li TT, Li J, Zuo AJ, Xu D, Song CX, Guo Y. CTRP9 induces iNOS expression through JAK2/STAT3 pathway in Raw 264.7 and peritoneal macrophages. Biochem Biophys Res Commun 2019; 523:98-104. [PMID: 31837806 DOI: 10.1016/j.bbrc.2019.12.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 12/02/2019] [Indexed: 01/10/2023]
Abstract
The C1q tumor necrosis factor (TNF)-related proteins 9 (CTRP9), an adipocyte-derived cytokine, affects a number of physiological processes, including immune function and inflammation. We investigated whether CTRP9 affects the expression of inflammation-related genes in Raw 264.7 and peritoneal macrophages. The CTRP9-induced expression of iNOS increased in a time- and dose-dependent manner. LPS and CTRP9 promote the expression of iNOS jointly in Raw 264.7 and peritoneal macrophages. CTRP9 induced the phosphorylation of JAK2 and STAT3 in Raw 264.7 and peritoneal macrophages. VX509 (JAK2 inhibitor) reduced the CTRP9-induced iNOS protein production. In addition, the CTRP9-induced phosphorylation of JAK2 and STAT3 was dramatically reduced by VX509. Collectively, these results suggest that JAK2/STAT3 signaling is involved in the CTRP9-induced expression of iNOS.
Collapse
Affiliation(s)
- Ji-Ying Chen
- Department of General Practice, Qilu Hospital of Shandong University, Jinan, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission, Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China
| | - Sheng-Yun Lei
- Department of General Practice, Qilu Hospital of Shandong University, Jinan, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission, Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China
| | - Ting-Ting Li
- Department of General Practice, Qilu Hospital of Shandong University, Jinan, China
| | - Jun Li
- Department of General Practice, Qilu Hospital of Shandong University, Jinan, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission, Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China
| | - An-Ju Zuo
- Department of General Practice, Qilu Hospital of Shandong University, Jinan, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission, Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China
| | - Dan Xu
- Department of General Practice, Qilu Hospital of Shandong University, Jinan, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission, Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China
| | - Cheng-Xiang Song
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission, Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yuan Guo
- Department of General Practice, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
30
|
The role of allogeneic stem-cell transplant in myelofibrosis in the era of JAK inhibitors: a case-based review. Bone Marrow Transplant 2019; 55:708-716. [PMID: 31534197 PMCID: PMC7113188 DOI: 10.1038/s41409-019-0683-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/29/2019] [Accepted: 08/02/2019] [Indexed: 01/11/2023]
Abstract
Allogeneic hematopoietic stem-cell transplantation (HSCT) is, at present, the only potentially curative therapy for myelofibrosis (MF). Despite many improvements, outcomes of HSCT are still burdened by substantial morbidity and high transplant-related mortality. Allogeneic transplant is generally considered in intermediate-2 and high-risk patients aged <70 years, but the optimal selection of patients and timing of the procedure remains under debate, as does as the role of JAK inhibitors in candidates for HSCT. Starting from a real-life clinical case scenario, herein we examine some of the crucial issues of HSCT for MF in light of recent refinements on MF risk stratification, data on the use of ruxolitinib before and after transplant and findings on the impact of different conditioning regimens and donor selection.
Collapse
|
31
|
Sailliet N, Brosseau C, Robert JM, Brouard S. Role of JAK inhibitors and immune cells in transplantation. Cytokine Growth Factor Rev 2019; 47:62-73. [DOI: 10.1016/j.cytogfr.2019.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 05/09/2019] [Indexed: 02/07/2023]
|
32
|
|
33
|
Bastian D, Wu Y, Betts BC, Yu XZ. The IL-12 Cytokine and Receptor Family in Graft-vs.-Host Disease. Front Immunol 2019; 10:988. [PMID: 31139181 PMCID: PMC6518430 DOI: 10.3389/fimmu.2019.00988] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 04/16/2019] [Indexed: 12/11/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is performed with curative intent for high- risk blood cancers and bone marrow failure syndromes; yet the development of acute and chronic graft-vs.-host disease (GVHD) remain preeminent causes of death and morbidity. The IL-12 family of cytokines is comprised of IL-12, IL-23, IL-27, IL-35, and IL-39. This family of cytokines is biologically distinct in that they are composed of functional heterodimers, which bind to cognate heterodimeric receptor chains expressed on T cells. Of these, IL-12 and IL-23 share a common β cytokine subunit, p40, as well as a receptor chain: IL-12Rβ1. IL-12 and IL-23 have been documented as proinflammatory mediators of GVHD, responsible for T helper 1 (Th1) differentiation and T helper 17 (Th17) stabilization, respectively. The role of IL-27 is less defined, seemingly immune suppressive via IL-10 secretion by Type 1 regulatory (Tr1) cells yet promoting inflammation through impairing CD4+ T regulatory (Treg) development and/or enhancing Th1 differentiation. More recently, IL-35 was described as a potent anti-inflammatory agent produced by regulatory B and T cells. The role of the newest member, IL-39, has been implicated in proinflammatory B cell responses but has not been explored in the context of allo-HCT. This review is directed at discussing the current literature relevant to each IL-12-family cytokine and cognate receptor engagement, as well as the consequential downstream signaling implications, during GVHD pathogenesis. Additionally, we will provide an overview of translational strategies targeting the IL-12 family cytokines, their receptors, and subsequent signal transduction to control GVHD.
Collapse
Affiliation(s)
- David Bastian
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Yongxia Wu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Brian C Betts
- Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Xue-Zhong Yu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
34
|
Betts BC, Locke FL, Sagatys EM, Pidala J, Walton K, Menges M, Reff J, Saha A, Djeu JY, Kiluk JV, Lee MC, Kim J, Kang CW, Tang CH, Frieling J, Lynch CC, List A, Rodriguez PC, Blazar BR, Conejo-Garcia JR, Del Valle JR, Hu CC, Anasetti C. Inhibition of Human Dendritic Cell ER Stress Response Reduces T Cell Alloreactivity Yet Spares Donor Anti-tumor Immunity. Front Immunol 2018; 9:2887. [PMID: 30574153 PMCID: PMC6291501 DOI: 10.3389/fimmu.2018.02887] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 11/26/2018] [Indexed: 11/13/2022] Open
Abstract
Acute graft- vs. -host disease (GVHD) is an important cause of morbidity and death after allogeneic hematopoietic cell transplantation (HCT). We identify a new approach to prevent GVHD that impairs monocyte-derived dendritic cell (moDC) alloactivation of T cells, yet preserves graft- vs.-leukemia (GVL). Exceeding endoplasmic reticulum (ER) capacity results in a spliced form of X-box binding protein-1 (XBP-1s). XBP-1s mediates ER stress and inflammatory responses. We demonstrate that siRNA targeting XBP-1 in moDCs abrogates their stimulation of allogeneic T cells. B-I09, an inositol-requiring enzyme-1α (IRE1α) inhibitor that prevents XBP-1 splicing, reduces human moDC migration, allo-stimulatory potency, and curtails moDC IL-1β, TGFβ, and p40 cytokines, suppressing Th1 and Th17 cell priming. B-I09-treated moDCs reduce responder T cell activation via calcium flux without interfering with regulatory T cell (Treg) function or GVL effects by cytotoxic T lymphocytes (CTL) and NK cells. In a human T cell mediated xenogeneic GVHD model, B-I09 inhibition of XBP-1s reduced target-organ damage and pathogenic Th1 and Th17 cells without impacting donor Tregs or anti-tumor CTL. DC XBP-1s inhibition provides an innovative strategy to prevent GVHD and retain GVL.
Collapse
Affiliation(s)
- Brian C Betts
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Tampa, FL, United States.,Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States.,Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, United States
| | - Frederick L Locke
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Tampa, FL, United States.,Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - Elizabeth M Sagatys
- Department of Hematopathology and Laboratory Medicine, Moffitt Cancer Center, Tampa, FL, United States
| | - Joseph Pidala
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Tampa, FL, United States.,Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - Kelly Walton
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States.,Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, United States
| | - Meghan Menges
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - Jordan Reff
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - Asim Saha
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States.,The Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Julie Y Djeu
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - John V Kiluk
- Comprehensive Breast Program, Moffitt Cancer Center, Tampa, FL, United States
| | - Marie C Lee
- Comprehensive Breast Program, Moffitt Cancer Center, Tampa, FL, United States
| | - Jongphil Kim
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, United States
| | - Chang Won Kang
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - Chih-Hang Tang
- Department of Translational Tumor Immunology, The Wistar Institute, Philadelphia, PA, United States
| | - Jeremy Frieling
- Department of Tumor Biology, Moffitt Cancer Center, Tampa, FL, United States
| | - Conor C Lynch
- Department of Tumor Biology, Moffitt Cancer Center, Tampa, FL, United States
| | - Alan List
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, United States
| | - Paulo C Rodriguez
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States.,The Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
| | | | - Juan R Del Valle
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - Chih-Chi Hu
- Department of Translational Tumor Immunology, The Wistar Institute, Philadelphia, PA, United States
| | - Claudio Anasetti
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Tampa, FL, United States.,Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
35
|
Iamsawat S, Daenthanasanmak A, Voss JH, Nguyen H, Bastian D, Liu C, Yu XZ. Stabilization of Foxp3 by Targeting JAK2 Enhances Efficacy of CD8 Induced Regulatory T Cells in the Prevention of Graft-versus-Host Disease. THE JOURNAL OF IMMUNOLOGY 2018; 201:2812-2823. [PMID: 30242073 DOI: 10.4049/jimmunol.1800793] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/31/2018] [Indexed: 02/02/2023]
Abstract
CD8+ induced regulatory T cells (iTregs) have been identified to suppress alloreactive immune responses and expressed regulatory T cell (Treg) ontological markers as similar as CD4+ iTregs. However, adoptive transfer of CD8+ iTreg-based therapy is hampered by the instability of Treg specific-transcription factor, Foxp3. As CD8+ iTregs were previously demonstrated to possess superior tumor-killing ability to CD4+ iTregs, adoptive transfer of stabilized CD8+ iTregs would be a potential therapy to prevent tumor relapse during graft-versus-leukemia disease (GVHD) treatment. In the current study, we generated alloantigen reactive CD8+ iTregs from JAK2-/- T cells and adoptively transferred them to MHC-mismatched and haploidentical murine models of allogeneic bone marrow transplantation. JAK2-/- CD8+ iTregs not only attenuated GVHD but also preserved graft-versus-leukemia effect. Mechanistic analysis revealed that JAK2-/- CD8+ iTregs upregulated natural Treg marker (neuropilin-1), and augmented DNA demethylation of CNS2 region within Foxp3 gene. These properties licensed JAK2-/- CD8+ iTregs to retain high Foxp3 expression resulting in less conversion to type 1 CTLs; as a result, JAK2-/- CD8+ iTregs were able to maintain their suppressive and cytolytic function. Thus, our findings provide a strong rationale and means to stabilize CD8+ iTregs by targeting JAK2, and the stabilized CD8+ iTregs exhibit therapeutic potential for alleviating GVHD and preserving the graft-versus-leukemia effect.
Collapse
Affiliation(s)
- Supinya Iamsawat
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425
| | - Anusara Daenthanasanmak
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425
| | - Jessica Heinrichs Voss
- 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
| | - David Bastian
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425
| | - Chen Liu
- Department of Pathology and Laboratory Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ 08901; and
| | - 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
|
36
|
Gadina M, Johnson C, Schwartz D, Bonelli M, Hasni S, Kanno Y, Changelian P, Laurence A, O'Shea JJ. Translational and clinical advances in JAK-STAT biology: The present and future of jakinibs. J Leukoc Biol 2018; 104:499-514. [PMID: 29999544 DOI: 10.1002/jlb.5ri0218-084r] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/24/2018] [Accepted: 05/28/2018] [Indexed: 02/06/2023] Open
Abstract
In this era, it is axiomatic that cytokines have critical roles in cellular development and differentiation, immune homeostasis, and host defense. Equally, dysregulation of cytokines is known to contribute to diverse inflammatory and immune-mediated disorders. In fact, the past 20 years have witnessed the rapid translation of basic discoveries in cytokine biology to multiple successful biological agents (mAbs and recombinant fusion proteins) that target cytokines. These targeted therapies have not only fundamentally changed the face of multiple immune-mediated diseases but have also unequivocally established the role of specific cytokines in human disease; cytokine biologists have many times over provided remarkable basic advances with direct clinical benefit. Numerous cytokines rely on the JAK-STAT pathway for signaling, and new, safe, and effective small molecule inhibitors have been developed for a range of disorders. In this review, we will briefly summarize basic discoveries in cytokine signaling and briefly comment on some major unresolved issues. We will review clinical data pertaining to the first generation of JAK inhibitors and their clinical indications, discuss additional opportunities for targeting this pathway, and lay out some of the challenges that lie ahead.
Collapse
Affiliation(s)
- Massimo Gadina
- National Institute of Arthritis, Musculoskeletal and Skin Diseases, Molecular Immunology and Inflammation Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Catrina Johnson
- National Institute of Arthritis, Musculoskeletal and Skin Diseases, Molecular Immunology and Inflammation Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Daniella Schwartz
- National Institute of Arthritis, Musculoskeletal and Skin Diseases, Molecular Immunology and Inflammation Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Michael Bonelli
- National Institute of Arthritis, Musculoskeletal and Skin Diseases, Molecular Immunology and Inflammation Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Sarfaraz Hasni
- National Institute of Arthritis, Musculoskeletal and Skin Diseases, Molecular Immunology and Inflammation Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Yuka Kanno
- National Institute of Arthritis, Musculoskeletal and Skin Diseases, Molecular Immunology and Inflammation Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Paul Changelian
- National Institute of Arthritis, Musculoskeletal and Skin Diseases, Molecular Immunology and Inflammation Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Arian Laurence
- National Institute of Arthritis, Musculoskeletal and Skin Diseases, Molecular Immunology and Inflammation Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - John J O'Shea
- National Institute of Arthritis, Musculoskeletal and Skin Diseases, Molecular Immunology and Inflammation Branch, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
37
|
Kröger N, Shahnaz Syed Abd Kadir S, Zabelina T, Badbaran A, Christopeit M, Ayuk F, Wolschke C. Peritransplantation Ruxolitinib Prevents Acute Graft-versus-Host Disease in Patients with Myelofibrosis Undergoing Allogenic Stem Cell Transplantation. Biol Blood Marrow Transplant 2018; 24:2152-2156. [PMID: 29800615 DOI: 10.1016/j.bbmt.2018.05.023] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 05/18/2018] [Indexed: 12/21/2022]
Abstract
JAK inhibition by ruxolitinib is approved for treating myelofibrosis and also has shown efficacy in treating steroid-resistant acute and chronic graft-versus-host disease (GVHD). In 12 patients with myelofibrosis (median age, 63 years; range, 43 to 71 years) who were treated with ruxolitinib and underwent allogeneic stem cell transplantation (ASCT), ruxolitinib was continued (2 × 5 mg daily) until stable engraftment. No graft failure was observed, and leukocyte engraftment was achieved after a median of 12 days (range, 11 to 18 days). One patient developed fever of unknown origin after discontinuation of ruxolitinib; otherwise, no withdrawal syndrome was observed. Overall, only 1 patient each experienced acute GVHD grade I or II, resulting in an 8% incidence of acute GVHD grade II-IV at day +100, with no nonrelapse mortality. Complete chimerism was achieved in 11 patients after a median of 40 days, and molecular clearance of the underlying driver mutation was noted in 10 patients after a median of 32 days. Cytomegalovirus (CMV) reactivation occurred in 5 patients (41%), 1 of whom had CMV colitis as well, but all resolved after ganciclovir treatment. In 2 patients, ruxolitinib had to be discontinued on day 17 and day 18 after ASCT due to cytopenia after engraftment. Levels of inflammatory cytokines IL-8, IL-10, IL-6, TNFR2, INF-α, and INF-β were reduced after ruxolitinib treatment. After day +100, 4 patients developed acute GVHD (1 with grade I, 2 with grade II, and 1 with grade III) after tapering of cyclosporine, and all patients were alive at a median follow-up of 17 months (range, 12 to 18 months).
Collapse
Affiliation(s)
- Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Sharifah Shahnaz Syed Abd Kadir
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Haematology, Ampang Hospital, Selangor, Malaysia
| | - Tatjana Zabelina
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anita Badbaran
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maximilian Christopeit
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Francis Ayuk
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christine Wolschke
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
38
|
Jagasia M, Zeiser R, Arbushites M, Delaite P, Gadbaw B, Bubnoff NV. Ruxolitinib for the treatment of patients with steroid-refractory GVHD: an introduction to the REACH trials. Immunotherapy 2018; 10:391-402. [PMID: 29316837 DOI: 10.2217/imt-2017-0156] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
For patients with hematologic malignancies and disorders, allogeneic hematopoietic stem cell transplantation offers a potentially curative treatment option. Many patients develop graft-versus-host disease (GVHD), a serious complication and leading cause of nonrelapse mortality. Corticosteroids are the standard first-line treatment for GVHD; however, patients often become steroid-refractory or remain corticosteroid-dependent. New second-line treatment options are needed to improve patient outcomes. Here we review the role of JAK1 and JAK2 in acute and chronic GVHD. We also describe the study designs of the Phase II REACH1 (NCT02953678) and the Phase III REACH2 (NCT02913261) and REACH3 (NCT03112603) clinical trials that are currently recruiting patients to evaluate the JAK1/JAK2 inhibitor ruxolitinib in patients with corticosteroid-refractory acute or chronic GVHD.
Collapse
Affiliation(s)
- Madan Jagasia
- Vanderbilt-Ingram Cancer Center, 1301 Medical Center Dr #1710, Nashville, TN, USA 37232
| | - Robert Zeiser
- Department of Hematology, Oncology & Stem Cell Transplantation, University Hospital Freiburg, Hugstetter Str. 55, 79106 Freiburg im Breisgau, Germany
| | | | - Patricia Delaite
- Incyte Corporation, 1801 Augustine Cut-off, Wilmington, DE, USA 19803
| | - Brian Gadbaw
- Novartis Pharmaceuticals Corporation, 1 Health Plaza, East Hanover, NJ, USA 07936
| | - Nikolas von Bubnoff
- Department of Hematology, Oncology & Stem Cell Transplantation, University Hospital Freiburg, Hugstetter Str. 55, 79106 Freiburg im Breisgau, Germany
| |
Collapse
|
39
|
MacDonald KPA, Betts BC, Couriel D. Reprint of: Emerging Therapeutics for the Control of Chronic Graft-versus-Host Disease. Biol Blood Marrow Transplant 2018; 24:S7-S14. [PMID: 29425517 DOI: 10.1016/j.bbmt.2017.12.788] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 10/03/2017] [Indexed: 02/02/2023]
Affiliation(s)
- Kelli P A MacDonald
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.
| | - Brian C Betts
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffit Cancer Center, Tampa, Florida
| | - Daniel Couriel
- Department of Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| |
Collapse
|
40
|
Targeting JAK2 reduces GVHD and xenograft rejection through regulation of T cell differentiation. Proc Natl Acad Sci U S A 2018; 115:1582-1587. [PMID: 29382747 DOI: 10.1073/pnas.1712452115] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Janus kinase 2 (JAK2) signal transduction is a critical mediator of the immune response. JAK2 is implicated in the onset of graft-versus-host disease (GVHD), which is a significant cause of transplant-related mortality after allogeneic hematopoietic cell transplantation (allo-HCT). Transfer of JAK2-/- donor T cells to allogeneic recipients leads to attenuated GVHD yet maintains graft-versus-leukemia. Th1 differentiation among JAK2-/- T cells is significantly decreased compared with wild-type controls. Conversely, iTreg and Th2 polarization is significantly increased among JAK2-/- T cells. Pacritinib is a multikinase inhibitor with potent activity against JAK2. Pacritinib significantly reduces GVHD and xenogeneic skin graft rejection in distinct rodent models and maintains donor antitumor immunity. Moreover, pacritinib spares iTregs and polarizes Th2 responses as observed among JAK2-/- T cells. Collectively, these data clearly identify JAK2 as a therapeutic target to control donor alloreactivity and promote iTreg responses after allo-HCT or solid organ transplantation. As such, a phase I/II acute GVHD prevention trial combining pacritinib with standard immune suppression after allo-HCT is actively being investigated (https://clinicaltrials.gov/ct2/show/NCT02891603).
Collapse
|
41
|
Betts BC, Veerapathran A, Pidala J, Yang H, Horna P, Walton K, Cubitt CL, Gunawan S, Lawrence HR, Lawrence NJ, Sebti SM, Anasetti C. Targeting Aurora kinase A and JAK2 prevents GVHD while maintaining Treg and antitumor CTL function. Sci Transl Med 2018; 9:9/372/eaai8269. [PMID: 28077684 DOI: 10.1126/scitranslmed.aai8269] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 08/17/2016] [Accepted: 10/10/2016] [Indexed: 12/12/2022]
Abstract
Graft-versus-host disease (GVHD) is a leading cause of nonrelapse mortality after allogeneic hematopoietic cell transplantation. T cell costimulation by CD28 contributes to GVHD, but prevention is incomplete when targeting CD28, downstream mammalian target of rapamycin (mTOR), or Aurora A. Likewise, interleukin-6 (IL-6)-mediated Janus kinase 2 (JAK2) signaling promotes alloreactivity, yet JAK2 inhibition does not eliminate GVHD. We provide evidence that blocking Aurora A and JAK2 in human T cells is synergistic in vitro, prevents xenogeneic GVHD, and maintains antitumor responses by cytotoxic T lymphocytes (CTLs). Aurora A/JAK2 inhibition is immunosuppressive but permits the differentiation of inducible regulatory T cells (iTregs) that are hyperfunctional and CD39 bright and efficiently scavenge adenosine triphosphate (ATP). Increased iTreg potency is primarily a function of Aurora A blockade, whereas JAK2 inhibition suppresses T helper 17 (TH17) differentiation. Inhibiting either Aurora A or JAK2 significantly suppresses TH1 T cells. However, CTL generated in vivo retains tumor-specific killing despite Aurora A/JAK2 blockade. Thus, inhibiting CD28 and IL-6 signal transduction pathways in donor T cells can increase the Treg/Tconv ratio, prevent GVHD, and preserve antitumor CTL.
Collapse
Affiliation(s)
- Brian C Betts
- Department of Blood and Marrow Transplantation, Moffitt Cancer Center, Tampa, FL 33612, USA. .,Department of Immunology, Moffitt Cancer Center, Tampa, FL 33612, USA.,Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA
| | - Anandharaman Veerapathran
- Department of Blood and Marrow Transplantation, Moffitt Cancer Center, Tampa, FL 33612, USA.,Department of Immunology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Joseph Pidala
- Department of Blood and Marrow Transplantation, Moffitt Cancer Center, Tampa, FL 33612, USA.,Department of Immunology, Moffitt Cancer Center, Tampa, FL 33612, USA.,Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA
| | - Hua Yang
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA
| | - Pedro Horna
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA.,Department of Hematopathology and Laboratory Medicine, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Kelly Walton
- Department of Immunology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | | | - Steven Gunawan
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA
| | - Harshani R Lawrence
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA.,Department of Drug Discovery, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Nicholas J Lawrence
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA.,Department of Drug Discovery, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Said M Sebti
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA.,Department of Drug Discovery, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Claudio Anasetti
- Department of Blood and Marrow Transplantation, Moffitt Cancer Center, Tampa, FL 33612, USA.,Department of Immunology, Moffitt Cancer Center, Tampa, FL 33612, USA.,Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
42
|
Hill L, Alousi A, Kebriaei P, Mehta R, Rezvani K, Shpall E. New and emerging therapies for acute and chronic graft versus host disease. Ther Adv Hematol 2018; 9:21-46. [PMID: 29317998 PMCID: PMC5753923 DOI: 10.1177/2040620717741860] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/10/2017] [Indexed: 12/14/2022] Open
Abstract
Graft versus host disease (GVHD) remains a major cause of morbidity and mortality following allogeneic hematopoietic stem-cell transplantation (HSCT). Despite the use of prophylactic GVHD regimens, a significant proportion of transplant recipients will develop acute or chronic GVHD following HSCT. Corticosteroids are standard first-line therapy, but are only effective in roughly half of all cases with ~50% of patients going on to develop steroid-refractory disease, which increases the risk of nonrelapse mortality. While progress has been made with improvements in survival outcomes over time, corticosteroids are associated with significant toxicities, and many currently available salvage therapies are associated with increased immunosuppression, infectious complications, and potential loss of the graft versus leukemia (GVL) effect. Thus, there is an unmet need for development of newer treatment strategies for both acute and chronic GVHD to improve long-term post-transplant outcomes and quality of life for HSCT recipients. Here, we provide a concise review of major emerging therapies currently being studied in the treatment of acute and chronic GVHD.
Collapse
Affiliation(s)
- LaQuisa Hill
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Amin Alousi
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer, Houston, TX, USA
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer, Houston, TX, USA
| | - Rohtesh Mehta
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer, Houston, TX, USA
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer, Houston, TX, USA
| | - Elizabeth Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 0423, Houston, TX 77030-4000, USA
| |
Collapse
|
43
|
Bai X, Shi H, Yang M, Wang Y, Sun Z, Xu S. Identification of key genes implicated in the suppressive function of human FOXP3+CD25+CD4+ regulatory T cells through the analysis of time‑series data. Mol Med Rep 2017; 17:3647-3657. [PMID: 29286140 PMCID: PMC5802170 DOI: 10.3892/mmr.2017.8366] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 04/27/2017] [Indexed: 01/01/2023] Open
Abstract
Human forkhead box P3 (FOXP3)+ cluster of differentiation (CD)25+CD4+ regulatory T cells (Tregs) are a type of T cell that express CD4, CD25 and FOXP3, which are critical for maintaining immune homeostasis. The present study aimed to determine the mechanisms underlying Treg function. The GSE11292 dataset was downloaded from the Gene Expression Omnibus, which included data from Treg cells at 19 time points (0–360 min) with an equal interval of 20 min, and corresponding repeated samples. However, data for Treg cells at time point 120 min were missing. Using the Mfuzz package, the key genes were identified by clustering analysis. Subsequently, regulatory networks and protein-protein interaction (PPI) networks were constructed and merged into integrated networks using Cytoscape software. Using Database for Annotation, Visualization and Integrated Discover software, enrichment analyses were performed for the genes involved in the PPI networks. Cluster 1 (including 292 genes), cluster 2 (including 111 genes), cluster 3 (including 194 genes) and cluster 4 (including 103 genes) were obtained from the clustering analysis. GAPDH (degree, 40) in cluster 1, Janus kinase 2 (JAK2) (degree, 10) and signal transducer and activator of transcription 5A (STAT5A) (degree, 9) in cluster 3, and tumor necrosis factor (TNF) (degree, 26) and interleukin 2 (IL2) (degree, 22) in cluster 4 had higher degrees in the PPI networks. In addition, it was indicated that several genes may have a role in Treg function by targeting other genes [e.g. microRNA (miR)-146b-3p→TNF, miR-146b-5p→TNF, miR-142-5p→TNF and tripartite motif containing 28 (TRIM28)→GAPDH]. Enrichment analyses indicated that IL2 and TNF were enriched in the immune response and T cell receptor signaling pathway. In conclusion, GAPDH targeted by TRIM28, TNF targeted by miR-146b-3p, miR-146b-5p and miR-142-5p, in addition to JAK2, IL2, and STAT5A may serve important roles in Treg function.
Collapse
Affiliation(s)
- Xiaofeng Bai
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Hua Shi
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Mingxi Yang
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Yuanlin Wang
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Zhaolin Sun
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Shuxiong Xu
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| |
Collapse
|
44
|
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: 65] [Impact Index Per Article: 9.3] [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
|
45
|
Affiliation(s)
- Robert Zeiser
- From the Department of Hematology, Oncology, and Stem Cell Transplantation, Faculty of Medicine, Freiburg University Medical Center, Freiburg, Germany (R.Z.); and the Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis (B.R.B.)
| | - Bruce R Blazar
- From the Department of Hematology, Oncology, and Stem Cell Transplantation, Faculty of Medicine, Freiburg University Medical Center, Freiburg, Germany (R.Z.); and the Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis (B.R.B.)
| |
Collapse
|
46
|
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: 1.7] [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
|
47
|
Emerging Therapeutics for the Control of Chronic Graft-versus-Host Disease. Biol Blood Marrow Transplant 2017; 24:19-26. [PMID: 29032060 DOI: 10.1016/j.bbmt.2017.10.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 10/03/2017] [Indexed: 02/01/2023]
|
48
|
Advances in the role of follicular T helper cells in graft versus host diseases. LIVER RESEARCH 2017. [DOI: 10.1016/j.livres.2017.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
49
|
Reinhardt-Heller K, Hirschberg I, Lang P, Vogl T, Handgretinger R, Bethge WA, Holzer U. Increase of Intermediate Monocytes in Graft-versus-Host Disease: Correlation with MDR1 +Th17.1 Levels and the Effect of Prednisolone and 1α,25-Dihydroxyvitamin D3. Biol Blood Marrow Transplant 2017; 23:2057-2064. [PMID: 28807771 DOI: 10.1016/j.bbmt.2017.08.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 08/03/2017] [Indexed: 11/30/2022]
Abstract
Graft-versus-host disease (GVHD) remains one of the major complications after allogeneic hematopoietic stem cell transplantation that is mainly treated with glucocorticoids such as prednisolone. In this study the influence of monocyte subpopulations, prednisolone, and 1α,25-dihydroxyvitamin D3 (1α,25-(OH)2D3) on the induction of a proinflammatory subset of Th17 cells (MDR+Th17.1) characterized by CCR6+CXCR3hiCCR4loCCR10-CD161+ and stable expression of the multidrug resistance protein type 1 (MDR1) was investigated. Our results demonstrate that intermediate monocytes are increased in patients with acute GVHD, promoting the induction of proinflammatory MDR1+Th17.1 cells. Furthermore, prednisolone induces the development of MDR1+Th17.1 cells, whereas 1α,25-(OH)2D3 acts as an anti-inflammatory, leading to diminished percentages of proinflammatory MDR1+Th17.1 cells in the presence of prednisolone after stimulation with the TLR4-ligand S100A8/S100A9. Moreover, 1α,25-(OH)2D3 decreased the expression level of the targets JAK2 and CD74, both associated with T cell activation, in monocytes. Thus, in steroid-resistant GVHD, 1α,25-(OH)2D3 could be an important regulator in monocyte-induced development of proinflammatory MDR1+Th17.1 cells and might therefore be a potential therapeutic agent in combination with glucocorticoids for GVHD treatment.
Collapse
Affiliation(s)
| | | | - Peter Lang
- Children's Hospital, University of Tuebingen, Germany
| | - Thomas Vogl
- Institute of Immunology, University of Muenster, Germany
| | | | | | - Ursula Holzer
- Children's Hospital, University of Tuebingen, Germany.
| |
Collapse
|
50
|
Tvedt THA, Ersvaer E, Tveita AA, Bruserud Ø. Interleukin-6 in Allogeneic Stem Cell Transplantation: Its Possible Importance for Immunoregulation and As a Therapeutic Target. Front Immunol 2017. [PMID: 28642760 PMCID: PMC5462914 DOI: 10.3389/fimmu.2017.00667] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Allogeneic stem cell transplantation is associated with a high risk of treatment-related mortality mainly caused by infections and graft-versus-host disease (GVHD). GVHD is characterized by severe immune dysregulation and impaired regeneration of different tissues, i.e., epithelial barriers and the liver. The balance between pro- and anti-inflammatory cytokine influences the risk of GVHD. Interleukin-6 (IL-6) is a cytokine that previously has been associated with pro-inflammatory effects. However, more recent evidence from various autoimmune diseases (e.g., inflammatory bowel disease, rheumatoid arthritis) has shown that the IL-6 activity is more complex with important effects also on tissue homeostasis, regeneration, and metabolism. This review summarizes the current understanding of how pro-inflammatory IL-6 effects exerted during the peritransplant period shapes T-cell polarization with enhancement of Th17 differentiation and suppression of regulatory T cells, and in addition we also review and discuss the results from trials exploring non-selective IL-6 inhibition in prophylaxis and treatment of GVHD. Emerging evidence suggests that the molecular strategy for targeting of IL-6-initiated intracellular signaling is important for the effect on GVHD. It will therefore be important to further characterize the role of IL-6 in the pathogenesis of GVHD to clarify whether combined IL-6 inhibition of both trans- (i.e., binding of the soluble IL-6/IL-6 receptor complex to cell surface gp130) and cis-signaling (i.e., IL-6 ligation of the IL-6 receptor/gp130 complex) or selective inhibition of trans-signaling should be tried in the prophylaxis and/or treatment of GVHD in allotransplant patients.
Collapse
Affiliation(s)
- Tor Henrik Anderson Tvedt
- Department of Clinical Science, Section for Hematology, University of Bergen, Bergen, Norway.,Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Elisabeth Ersvaer
- Institute of Biomedical Laboratory Sciences and Chemical Engineering, Western Norway University of Applied Sciences (HVL), Bergen, Norway
| | - Anders Aune Tveita
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, Oslo, Norway
| | - Øystein Bruserud
- Department of Clinical Science, Section for Hematology, University of Bergen, Bergen, Norway.,Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|