1
|
Yang PJ, Zhao XY, Kou YH, Liu J, Ren XY, Zhang YY, Wang ZD, Ge Z, Yuan WX, Qiu C, Tan B, Liu Q, Shi YN, Jiang YQ, Qiu C, Guo LH, Li JY, Huang XJ, Yu LY. Human amniotic epithelial stem cell is a cell therapy candidate for preventing acute graft-versus-host disease. Acta Pharmacol Sin 2024; 45:2339-2353. [PMID: 38802569 PMCID: PMC11489431 DOI: 10.1038/s41401-024-01283-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 04/01/2024] [Indexed: 05/29/2024] Open
Abstract
Graft-versus-host disease (GVHD), an immunological disorder that arises from donor T cell activation through recognition of host alloantigens, is the major limitation in the application of allogeneic hematopoietic stem cell transplantation (allo-HSCT). Traditional immunosuppressive agents can relieve GVHD, but they induce serious side effects. It is highly required to explore alternative therapeutic strategy. Human amniotic epithelial stem cells (hAESCs) were recently considered as an ideal source for cell therapy with special immune regulatory property. In this study, we evaluated the therapeutic role of hAESCs in the treatment of GVHD, based on our previous developed cGMP-grade hAESCs product. Humanized mouse model of acute GVHD (aGVHD) was established by injection of huPBMCs via the tail vein. For prevention or treatment of aGVHD, hAESCs were injected to the mice on day -1 or on day 7 post-PBMC infusion, respectively. We showed that hAESCs infusion significantly alleviated the disease phenotype, increased the survival rate of aGVHD mice, and ameliorated pathological injuries in aGVHD target organs. We demonstrated that hAESCs directly induced CD4+ T cell polarization, in which Th1 and Th17 subsets were downregulated, and Treg subset was elevated. Correspondingly, the levels of a series of pro-inflammatory cytokines were reduced while the levels of the anti-inflammatory cytokines were upregulated in the presence of hAESCs. We found that hAESCs regulated CD4+ subset polarization in a paracrine mode, in which TGFβ and PGE2 were selectively secreted to mediate Treg elevation and Th1/Th17 inhibition, respectively. In addition, transplanted hAESCs preserved the graft-versus-leukemia (GVL) effect by inhibiting leukemia cell growth. More intriguingly, hAESCs infusion in HSCT patients displayed potential anti-GVHD effect with no safety concerns and confirmed the immunoregulatory mechanisms in the preclinical study. We conclude that hAESCs infusion is a promising therapeutic strategy for post-HSCT GVHD without compromising the GVL effect. The clinical trial was registered at www.clinicaltrials.gov as #NCT03764228.
Collapse
Affiliation(s)
- Peng-Jie Yang
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310058, China
| | - Xiang-Yu Zhao
- Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University, Beijing, 100044, China
| | - Yao-Hui Kou
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310058, China
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, 314400, China
| | - Jia Liu
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310058, China
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, 314400, China
| | - Xiang-Yi Ren
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310058, China
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, 314400, China
| | - Yuan-Yuan Zhang
- Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University, Beijing, 100044, China
| | - Zhi-Dong Wang
- Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University, Beijing, 100044, China
| | - Zhen Ge
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 310013, China
| | - Wei-Xin Yuan
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, 314400, China
- Shanghai iCELL Biotechnology Co. Ltd, Shanghai, 200335, China
| | - Chen Qiu
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310058, China
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, 314400, China
| | - Bing Tan
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310058, China
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, 314400, China
| | - Qin Liu
- Shanghai iCELL Biotechnology Co. Ltd, Shanghai, 200335, China
| | - Yan-Na Shi
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, 314400, China
| | - Yuan-Qing Jiang
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310058, China
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, 314400, China
| | - Cong Qiu
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310058, China
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, 314400, China
| | - Li-He Guo
- Shanghai iCELL Biotechnology Co. Ltd, Shanghai, 200335, China
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jin-Ying Li
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310058, China.
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, 314400, China.
| | - Xiao-Jun Huang
- Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University, Beijing, 100044, China.
- Peking-Tsinghua Center for Life Sciences, Beijing, 100084, China.
| | - Lu-Yang Yu
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310058, China.
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, 314400, China.
| |
Collapse
|
2
|
Chen Y, Feng J, Chen Y, Xia C, Yao M, Ding W, Li X, Fu X, Zheng S, Ma Y, Zou J, Lan M, Gao F. ROS-responsive nano-medicine for navigating autophagy to enhance targeted therapy of inflammatory bowel disease. Int J Pharm 2024; 659:124117. [PMID: 38615805 DOI: 10.1016/j.ijpharm.2024.124117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 04/16/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic gastrointestinal disorder characterized by immune dysregulation and intestinal inflammation. Rapamycin (Ra), an mTORC1 pathway inhibitor, has shown promise for autophagy induction in IBD therapy but is associated with off-target effects and toxicity. To address these issues, we developed an oral liposome responsive to reactive oxygen species (ROS) using lipids and amphiphilic materials. We combined ketone thiol (TK) for ROS responsive and hyaluronic acid (HA) with high affinity for CD44 receptors to prepare rapamycin-loaded nanoparticle (Ra@TH). Owing to its ROS responsive characteristic, Ra@TH can achieve inflammatory colonic targeting. Additionally, Ra@TH can induce autophagy by inhibiting the mTORC1 pathway, leading to the clearance of damaged organelles, pathogenic microorganisms and oxidative stress products. Simultaneously, it also collaboratively inhibits the NF-κB pathway suppressed by the removal of ROS resulting from TK cleavage, thereby mediating the expression of inflammatory factors. Furthermore, Ra@TH enhances the expression of typical tight junction proteins, synergistically restoring intestinal barrier function. Our research not only expands the understanding of autophagy in IBD treatment but also introduces a promising therapeutic approach for IBD patients.
Collapse
Affiliation(s)
- You Chen
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Juewen Feng
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yang Chen
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Chuanhe Xia
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Min Yao
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Wenxing Ding
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Xiang Li
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Xiuzhi Fu
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Shulei Zheng
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yin Ma
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Jiafeng Zou
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China; Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; Shanghai Key Laboratory of New Drug Design, East China University of Science and Technology, Shanghai 200237, China
| | - Minbo Lan
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China
| | - Feng Gao
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China; Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; Shanghai Key Laboratory of New Drug Design, East China University of Science and Technology, Shanghai 200237, China; Optogenetics and Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
3
|
Kim DDH, Popradi G, Lepic K, Paulson K, Allan D, Nampoothiri RV, Lachance S, Deotare U, White J, Elemary M, Jamani K, Fraga C, Lemieux C, Novitzky-Basso I, Law AD, Kumar R, Walker I, Schultz KR. Cell Therapy Transplant Canada (CTTC) Consensus-Based Guideline 2024 for Management and Treatment of Chronic Graft-Versus-Host Disease and Future Directions for Development. Curr Oncol 2024; 31:1426-1444. [PMID: 38534941 PMCID: PMC10968999 DOI: 10.3390/curroncol31030108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 01/28/2024] [Accepted: 02/28/2024] [Indexed: 05/26/2024] Open
Abstract
This is a consensus-based Canadian guideline whose primary purpose is to standardize and facilitate the management of chronic graft-versus-host disease (cGvHD) across the country. Creating uniform healthcare guidance in Canada is a challenge for a number of reasons including the differences in healthcare authority structure, funding and access to healthcare resources between provinces and territories, as well as the geographic size. These differences can lead to variable and unequal access to effective therapies for GvHD. This document will provide comprehensive and practical guidance that can be applied across Canada by healthcare professionals caring for patients with cGvHD. Hopefully, this guideline, based on input from GvHD treaters across the country, will aid in standardizing cGvHD care and facilitate access to much-needed novel therapies. This consensus paper aims to discuss the optimal approach to the initial assessment of cGvHD, review the severity scoring and global grading system, discuss systemic and topical treatments, as well as supportive therapies, and propose a therapeutic algorithm for frontline and subsequent lines of cGvHD treatment in adults and pediatric patients. Finally, we will make suggestions about the future direction of cGvHD treatment development such as (1) a mode-of-action-based cGvHD drug selection, according to the pathogenesis of cGvHD, (2) a combination strategy with the introduction of newer targeted drugs, (3) a steroid-free regimen, particularly for front line therapy for cGvHD treatment, and (4) a pre-emptive approach which can prevent the progression of cGvHD in high-risk patients destined to develop severe and highly morbid forms of cGvHD.
Collapse
Affiliation(s)
- Dennis Dong Hwan Kim
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 2M9, Canada (A.D.L.)
| | - Gizelle Popradi
- Health Centre, McGill University, Montreal, QC H4P 2P5, Canada
| | - Kylie Lepic
- Juravinski Cancer Centre, McMaster University, Hamilton, ON L8V 5C2, Canada
| | - Kristjan Paulson
- CancerCare Manitoba, University of Manitoba, Winnipeg, Manitoba, MB R3E 0V9, Canada
| | - David Allan
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON K1H 8L6, Canada
| | | | - Sylvie Lachance
- Hôpital Maisonneuve-Rosemont, Department of Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada;
| | - Uday Deotare
- London Health Sciences Centre, University of Western Ontario, London, ON N6A 5A5, Canada
| | - Jennifer White
- Vancouver General Hospital, British Columbia Cancer Agency, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Mohamed Elemary
- Saskatchewan Cancer Agency, University of Saskatchewan, Saskatchewan, SK S7N 4H4, Canada
| | - Kareem Jamani
- Tom Baker Cancer Centre, University of Calgary, Calgary, AB T2N 4N2, Canada;
| | - Christina Fraga
- Queen Elizabeth II Health Sciences Centre, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Christopher Lemieux
- CHU de Québec—Université Laval, Université Laval, Québec, QC G1R 2J6, Canada
| | - Igor Novitzky-Basso
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 2M9, Canada (A.D.L.)
| | - Arjun Datt Law
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 2M9, Canada (A.D.L.)
| | - Rajat Kumar
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 2M9, Canada (A.D.L.)
| | - Irwin Walker
- Juravinski Cancer Centre, McMaster University, Hamilton, ON L8V 5C2, Canada
| | - Kirk R. Schultz
- British Columbia Children’s Hospital, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| |
Collapse
|
4
|
Ling B, Xu Y, Qian S, Xiang Z, Xuan S, Wu J. Regulation of hematopoietic stem cells differentiation, self-renewal, and quiescence through the mTOR signaling pathway. Front Cell Dev Biol 2023; 11:1186850. [PMID: 37228652 PMCID: PMC10203478 DOI: 10.3389/fcell.2023.1186850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/28/2023] [Indexed: 05/27/2023] Open
Abstract
Hematopoietic stem cells (HSCs) are important for the hematopoietic system because they can self-renew to increase their number and differentiate into all the blood cells. At a steady state, most of the HSCs remain in quiescence to preserve their capacities and protect themselves from damage and exhaustive stress. However, when there are some emergencies, HSCs are activated to start their self-renewal and differentiation. The mTOR signaling pathway has been shown as an important signaling pathway that can regulate the differentiation, self-renewal, and quiescence of HSCs, and many types of molecules can regulate HSCs' these three potentials by influencing the mTOR signaling pathway. Here we review how mTOR signaling pathway regulates HSCs three potentials, and introduce some molecules that can work as the regulator of HSCs' these potentials through the mTOR signaling. Finally, we outline the clinical significance of studying the regulation of HSCs three potentials through the mTOR signaling pathway and make some predictions.
Collapse
Affiliation(s)
- Bai Ling
- Department of Pharmacy, The Yancheng Clinical College of Xuzhou Medical University, The First People’s Hospital of Yancheng, Yancheng, Jiangsu, China
| | - Yunyang Xu
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Siyuan Qian
- The Second School of Clinical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Ze Xiang
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shihai Xuan
- Department of Laboratory Medicine, The People’s Hospital of Dongtai City, Dongtai, China
| | - Jian Wu
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| |
Collapse
|
5
|
Villa A, Kuten-Shorrer M. Pathogenesis of Oral Toxicities Associated with Targeted Therapy and Immunotherapy. Int J Mol Sci 2023; 24:ijms24098188. [PMID: 37175898 PMCID: PMC10179284 DOI: 10.3390/ijms24098188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/04/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
Targeted therapy and immunotherapy have redefined cancer treatment. While they have enhanced tumor response and improved survival rates in many cancer types, toxicities continue to occur, and these often involve the oral cavity. Broadly reported as "mucositis" or "stomatitis," oral toxicities induced by targeted therapies differ clinically and mechanistically from those associated with conventional chemotherapy. Manifesting primarily as mucosal lesions, salivary gland hypofunction, or orofacial neuropathies, these oral toxicities may nonetheless lead to significant morbidity and impact patients' quality of life, thereby compromising clinical outcomes. We conclude that familiarity with the spectrum of associated toxicities and understanding of their pathogenesis represent important areas of clinical research and may lead to better characterization, prevention, and management of these adverse events.
Collapse
Affiliation(s)
- Alessandro Villa
- Oral Medicine, Oral Oncology and Dentistry, Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA
- The Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33176, USA
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Michal Kuten-Shorrer
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, New York, NY 14642, USA
| |
Collapse
|
6
|
Song Q, Nasri U, Nakamura R, Martin PJ, Zeng D. Retention of Donor T Cells in Lymphohematopoietic Tissue and Augmentation of Tissue PD-L1 Protection for Prevention of GVHD While Preserving GVL Activity. Front Immunol 2022; 13:907673. [PMID: 35677056 PMCID: PMC9168269 DOI: 10.3389/fimmu.2022.907673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 04/26/2022] [Indexed: 11/30/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (Allo-HCT) is a curative therapy for hematological malignancies (i.e., leukemia and lymphoma) due to the graft-versus-leukemia (GVL) activity mediated by alloreactive T cells that can eliminate residual malignant cells and prevent relapse. However, the same alloreactive T cells can cause a serious side effect, known as graft-versus-host disease (GVHD). GVHD and GVL occur in distinct organ and tissues, with GVHD occurring in target organs (e.g., the gut, liver, lung, skin, etc.) and GVL in lympho-hematopoietic tissues where hematological cancer cells primarily reside. Currently used immunosuppressive drugs for the treatment of GVHD inhibit donor T cell activation and expansion, resulting in a decrease in both GVHD and GVL activity that is associated with cancer relapse. To prevent GVHD, it is important to allow full activation and expansion of alloreactive T cells in the lympho-hematopoietic tissues, as well as prevent donor T cells from migrating into the GVHD target tissues, and tolerize infiltrating T cells via protective mechanisms, such as PD-L1 interacting with PD-1, in the target tissues. In this review, we will summarize major approaches that prevent donor T cell migration into GVHD target tissues and approaches that augment tolerization of the infiltrating T cells in the GVHD target tissues while preserving strong GVL activity in the lympho-hematopoietic tissues.
Collapse
Affiliation(s)
- Qingxiao Song
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, Unites States.,Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, Unites States.,Fujian Medical University Center of Translational Hematology, Fujian Institute of Hematology, and Fujian Medical University Union Hospital, Fuzhou, China
| | - Ubaydah Nasri
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, Unites States.,Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, Unites States
| | - Ryotaro Nakamura
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, Unites States
| | - Paul J Martin
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA, United States
| | - Defu Zeng
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, Unites States.,Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, Unites States
| |
Collapse
|
7
|
Mirza AS, Tandon A, Jenneman D, Cao S, Brimer T, Kumar A, Kidd M, Khimani F, Faramand R, Mishra A, Liu H, Nishihori T, Perez L, Lazaryan A, Bejanyan N, Nieder M, Pidala J, Elmariah H. Outcomes Following Intolerance to Tacrolimus/Sirolimus Graft-Versus-Host Disease Prophylaxis for Allogeneic Hematopoietic Cell Transplantation. Transplant Cell Ther 2022; 28:185.e1-185.e7. [PMID: 35017119 DOI: 10.1016/j.jtct.2022.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/07/2021] [Accepted: 01/04/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Although tacrolimus and sirolimus (TAC/SIR) is an accepted graft-versus-host disease (GVHD) prophylaxis following allogeneic hematopoietic cell transplant (HCT), toxicity from this regimen can lead to premature discontinuation of immunosuppression. There are limited studies reporting outcomes and subsequent treatment of patients with TAC/SIR intolerance. OBJECTIVES To assess outcomes of patients with TAC/SIR intolerance and guide subsequent management after intolerance. STUDY DESIGN We retrospectively analyzed transplant outcomes of consecutive adult patients at Moffitt Cancer Center who received allogeneic HCT with TAC/SIR as GVHD prophylaxis from 2009 to 2018. TAC/SIR intolerance was defined as discontinuation due to toxicity of either TAC or SIR before post-transplant day 100. RESULTS 777 patients met the inclusion criteria. Median follow-up was 22 (0.2-125) months. Intolerance occurred in 13% (n = 104) of patients at a median of 30 (range 5-90) days. The most common causes of intolerance were acute kidney injury (n = 53 [51%]), thrombotic microangiopathy (n = 31 [28%]), and veno-occlusive disease (n = 23 [22%]). The cumulative incidence of grade 2 to 4 acute GVHD at 100 days in TAC/SIR-intolerant patients was 50% (95% CI, 39%-64%) and 25% (95% CI, 22%-29%) in patients tolerant to this regimen (P < .0001). In multivariate analyses, grade 2 to 4 acute GVHD was significantly higher in TAC/SIR-intolerant patients (HR 2.40; 95% CI, 1.59-3.61; P < .0001). Similarly, in multivariate analyses, TAC/SIR-intolerant patients had more chronic GVHD (HR 1.48, 95% CI, 1.03-2.12; P = .03). The non-relapse mortality (NRM) at 1 year in TAC/SIR-intolerant patients was 47% (95% CI, 38%-59%) and 12% (95% CI, 10%-15%) in those tolerant to the regimen (P < .0001). The 2-year relapse free survival of TAC/SIR-intolerant patients was 35% (95% CI, 25%-44%) and 60% (95% CI, 57%-65%) among TAC/SIR-tolerant patients, (HR 2.30; 95% CI, 1.61-3.28; P < .0001). Intolerance stratified by early (≤30 days) versus late (31-100 days) significantly affected the cumulative incidence of acute GVHD at 75% (early [95% CI, 59%-94%]) versus 33% ([late] 95% CI, 21%-50%) (P = .001) as well as the cumulative incidence of NRM at 61% ([early] 95% CI, 48%-77%) versus 35% ([late] 95% CI, 24%-51%) (P = .006). After developing TAC/SIR intolerance, most patients were switched to an alternative 2-drug regimen (71/104 [68%]), with the most common being mycophenolate mofetil in addition to continuing TAC or SIR (68/71 [96%]). CONCLUSIONS Overall, TAC/SIR intolerance was associated with poorer outcomes. Early intolerance contributed to higher risk of acute GVHD, increased NRM, and inferior survival. Patients with early intolerance were often switched to an alternative agent, and patients with late intolerance tended to be continued on single-drug therapy without substitution. Single-drug versus 2-drug regimens after intolerance did not appear to affect outcomes. Management strategies to mitigate the risks of intolerance are warranted.
Collapse
Affiliation(s)
- Abu-Sayeef Mirza
- University of South Florida, Dept of Internal Medicine, Tampa, FL
| | - Ankita Tandon
- University of South Florida, Dept of Internal Medicine, Tampa, FL
| | - Dakota Jenneman
- University of South Florida, Dept of Internal Medicine, Tampa, FL
| | - Shu Cao
- University of South Florida, Dept of Internal Medicine, Tampa, FL
| | - Thomas Brimer
- University of South Florida, Dept of Internal Medicine, Tampa, FL
| | - Ambuj Kumar
- Morsani College of Medicine, Dept of Evidence Based Medicine, Tampa FL
| | - Michelle Kidd
- Moffitt Cancer Center, Dept. of Blood & Marrow Transplant and Cellular Immunotherapy, Tampa, FL
| | - Farhad Khimani
- Moffitt Cancer Center, Dept. of Blood & Marrow Transplant and Cellular Immunotherapy, Tampa, FL
| | - Rawan Faramand
- Moffitt Cancer Center, Dept. of Blood & Marrow Transplant and Cellular Immunotherapy, Tampa, FL
| | - Asmita Mishra
- Moffitt Cancer Center, Dept. of Blood & Marrow Transplant and Cellular Immunotherapy, Tampa, FL
| | - Hien Liu
- Moffitt Cancer Center, Dept. of Blood & Marrow Transplant and Cellular Immunotherapy, Tampa, FL
| | - Taiga Nishihori
- Moffitt Cancer Center, Dept. of Blood & Marrow Transplant and Cellular Immunotherapy, Tampa, FL
| | - Lia Perez
- Moffitt Cancer Center, Dept. of Blood & Marrow Transplant and Cellular Immunotherapy, Tampa, FL
| | - Aleksandr Lazaryan
- Moffitt Cancer Center, Dept. of Blood & Marrow Transplant and Cellular Immunotherapy, Tampa, FL
| | - Nelli Bejanyan
- Moffitt Cancer Center, Dept. of Blood & Marrow Transplant and Cellular Immunotherapy, Tampa, FL
| | - Michael Nieder
- Moffitt Cancer Center, Dept. of Blood & Marrow Transplant and Cellular Immunotherapy, Tampa, FL
| | - Joseph Pidala
- Moffitt Cancer Center, Dept. of Blood & Marrow Transplant and Cellular Immunotherapy, Tampa, FL
| | - Hany Elmariah
- Moffitt Cancer Center, Dept. of Blood & Marrow Transplant and Cellular Immunotherapy, Tampa, FL.
| |
Collapse
|
8
|
Sobkowiak-Sobierajska A, Lindemans C, Sykora T, Wachowiak J, Dalle JH, Bonig H, Gennery A, Lawitschka A. Management of Chronic Graft-vs.-Host Disease in Children and Adolescents With ALL: Present Status and Model for a Personalised Management Plan. Front Pediatr 2022; 10:808103. [PMID: 35252060 PMCID: PMC8894895 DOI: 10.3389/fped.2022.808103] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/24/2022] [Indexed: 12/18/2022] Open
Abstract
Herein we review current practice regarding the management of chronic graft-vs.-host disease (cGvHD) in paediatric patients after allogeneic haematopoietic stem cell transplantation (HSCT) for acute lymphoblastic leukaemia (ALL). Topics covered include: (i) the epidemiology of cGvHD; (ii) an overview of advances in our understanding cGvHD pathogenesis; (iii) current knowledge regarding risk factors for cGvHD and prevention strategies complemented by biomarkers; (iii) the paediatric aspects of the 2014 National Institutes for Health-defined diagnosis and grading of cGvHD; and (iv) current options for cGvHD treatment. We cover topical therapy and newly approved tyrosine kinase inhibitors, emphasising the use of immunomodulatory approaches in the context of the delicate counterbalance between immunosuppression and immune reconstitution as well as risks of relapse and infectious complications. We examine real-world approaches of response assessment and tapering schedules of treatment. Furthermore, we report on the optimal timepoints for therapeutic interventions and changes in relation to immune reconstitution and risk of relapse/infection. Additionally, we review the different options for anti-infectious prophylaxis. Finally, we put forth a theory of a holistic view of paediatric cGvHD and its associated manifestations and propose a checklist for individualised risk evaluation with aggregated considerations including site-specific cGvHD evaluation with attention to each individual's GvHD history, previous medical history, comorbidities, and personal tolerance and psychosocial circumstances. To complement this checklist, we present a treatment algorithm using representative patients to inform the personalised management plans for patients with cGvHD after HSCT for ALL who are at high risk of relapse.
Collapse
Affiliation(s)
| | - Caroline Lindemans
- Department of Pediatrics, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Pediatric Blood and Bone Marrow Transplantation, Princess Máxima Center, Utrecht, Netherlands
| | - Tomas Sykora
- Department of Pediatric Hematology and Oncology - Haematopoietic Stem Cell Transplantation Unit, National Institute of Children's Diseases and Medical Faculty, Comenius University, Bratislava, Slovakia
| | - Jacek Wachowiak
- Department of Pediatric Oncology, Hematology and Transplantology, Poznan University of Medical Sciences, Poznan, Poland
| | - Jean-Hugues Dalle
- Hematology and Immunology Department, Robert-Debré Hospital, Assistance Publique-Hôpitaux de Paris and University of Paris, Paris, France
| | - Halvard Bonig
- Goethe University Medical Center, Institute of Transfusion Medicine and Immunohematology, and German Red Cross Blood Center Frankfurt, Frankfurt, Germany
| | - Andrew Gennery
- Medical School, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Anita Lawitschka
- Stem Cell Transplantation Unit, St. Anna Children's Hospital, Medical University Vienna, Vienna, Austria.,St. Anna Children's Cancer Research Institute, Vienna, Austria
| |
Collapse
|
9
|
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: 26] [Impact Index Per Article: 6.5] [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
|
10
|
Demosthenous C, Sakellari I, Douka V, Papayanni PG, Anagnostopoulos A, Gavriilaki E. The Role of Myeloid-Derived Suppressor Cells (MDSCs) in Graft-versus-Host Disease (GVHD). J Clin Med 2021; 10:jcm10102050. [PMID: 34064671 PMCID: PMC8150814 DOI: 10.3390/jcm10102050] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/02/2021] [Accepted: 05/06/2021] [Indexed: 12/11/2022] Open
Abstract
Background: Myeloid-derived suppressor cells (MDSCs) are implicated in the complex interplay involving graft-versus-leukemia (GVL) effects and graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (allo-HCT) in hematologic malignancies. Methods: A review of literature through PubMed was undertaken to summarize the published evidence on the pathophysiology and clinical implications of MDSCs in allo-HCT. Literature sources published in English since 1978 were searched, using the terms Natural Suppressor (NS) cells, MDSCs, GVHD, and allo-HCT. Results: In vivo studies demonstrated that MDSCs derived from mobilization protocols could strongly suppress allo-responses mediated by T cells and enhance T-Reg activity, thus inhibiting GVHD toxicity. However, the influence of MDSCs on the GVL effect is not fully defined. Conclusions: The induction or maintenance of MDSC suppressive function would be advantageous in suppressing inflammation associated with GVHD. Pathways involved in MDSC metabolism and the inflammasome signaling are a promising field of study to elucidate the function of MDSCs in the pathogenesis of GVHD and translate these findings to a clinical setting.
Collapse
|
11
|
Kanunnikov MM, Rakhmanova ZZ, Levkovsky NV, Vafina AI, Goloshapov OV, Shchegoleva TS, Vlasova JJ, Paina OV, Morozova EV, S Zubarovskaya L, Kulagin AD, S Moiseev I. Conversion from calcineurin inhibitors to sirolimus in transplant-associated thrombotic microangiopathy. Clin Transplant 2020; 35:e14180. [PMID: 33258122 DOI: 10.1111/ctr.14180] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/18/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022]
Abstract
Transplant-associated thrombotic microangiopathy (TA-TMA) is a specific complication of allogeneic hematopoietic cell transplantation with a multifactorial etiology. There is little evidence published regarding the efficacy and factors influencing the outcome of substitution of calcineurin inhibitors (CNIs) with other agentsas a widely accepted practice in this disorder; however, there are limited data on the options for immunosuppression manipulation (ISM). In our study, we retrospectively analyzed outcomes of 45 patients with TA-TMA with ISM and substitution either with steroids (steroid group) or anmTOR inhibitor sirolimus (sirolimus group). In our study, sirolimus was associated with significantly better 1-year overall survival (HR 0.3, 95% CI 0.13-0.7, p = .004) and faster time to normalization of LDH (HR 2.2, 95% CI 0.99-4.99, p = .044). Replacing CNIs with sirolimus could be an effective option in patients with TA-TMA. A multicenter confirmatory study of CNIs replacement with sirolimus is justified.
Collapse
Affiliation(s)
| | | | - Nikita V Levkovsky
- RM Gorbacheva Research Institute, Pavlov University, St. Petersburg, Russia
| | - Aliya I Vafina
- RM Gorbacheva Research Institute, Pavlov University, St. Petersburg, Russia
| | - Oleg V Goloshapov
- RM Gorbacheva Research Institute, Pavlov University, St. Petersburg, Russia
| | | | - Julia J Vlasova
- RM Gorbacheva Research Institute, Pavlov University, St. Petersburg, Russia
| | - Olesya V Paina
- RM Gorbacheva Research Institute, Pavlov University, St. Petersburg, Russia
| | - Elena V Morozova
- RM Gorbacheva Research Institute, Pavlov University, St. Petersburg, Russia
| | | | | | - Ivan S Moiseev
- RM Gorbacheva Research Institute, Pavlov University, St. Petersburg, Russia
| |
Collapse
|
12
|
Gawedzki P, Collins J. Impact of the implementation of a pharmacist-driven immunosuppression drug monitoring protocol for hematopoietic stem cell transplant recipients. J Oncol Pharm Pract 2020; 27:1907-1913. [PMID: 33250016 DOI: 10.1177/1078155220975088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
For patients with serious hematologic malignancies, hematopoietic stem cell transplantation (HSCT) is a potentially curative treatment option. Majority of HSCT recipients receive tacrolimus as part of their immunosuppressive regimen. The purpose of this study is to evaluate the clinical impact of a pharmacist driven immunosuppression drug monitoring protocol for HSCT recipients on tacrolimus.This was a single-center, pre-post interventional study conducted at the University of Chicago Medical Center. Data collected via chart review includes the immunosuppressive agent used, interacting medications, adverse events, dose adjustments, drug concentrations, time to engraftment, and diagnosis of GVHD.Following the incorporation of a therapeutic drug monitoring protocol, the percentage of therapeutic tacrolimus levels was similar to when there was no protocol in place; 68% versus 64%, respectively (p = 0.34). There were 18 total adverse events observed in the pre-protocol group versus 10 in the post-protocol group (p = 0.03). Nephrotoxicity was the most common adverse event occurring in 23% of patients in the pre-protocol group and 15% of patients in the post-protocol group (p = 0.18). In the post-protocol group, there were 20 patients with two or more interacting drugs versus two patients in the pre-protocol group (p < 0.05). Additionally, the post-protocol group had 12 instances of an empiric dose adjustment made whereas the pre-protocol group had three instances (p = 0.006).Although there was no significant difference in percentage of therapeutic tacrolimus levels, pharmacist involvement resulted in improved safety outcomes such as management of drug interactions and incidence of adverse events.
Collapse
Affiliation(s)
- Paula Gawedzki
- Department of Pharmacy Services, 2462University of Chicago Medicine, Chicago, IL, USA
| | - Jennifer Collins
- Department of Pharmacy Services, 2462University of Chicago Medicine, Chicago, IL, USA
| |
Collapse
|
13
|
Brown RA, Byersdorfer CA. Metabolic Pathways in Alloreactive T Cells. Front Immunol 2020; 11:1517. [PMID: 32793207 PMCID: PMC7393946 DOI: 10.3389/fimmu.2020.01517] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (aHSCT) is a curative therapy for a range of hematologic illnesses including aplastic anemia, sickle cell disease, immunodeficiency, and high-risk leukemia, but the efficacy of aHSCT is often undermined by graft-versus-host disease (GVHD), where T cells from the donor attack and destroy recipient tissues. Given the strong interconnection between T cell metabolism and cellular function, determining the metabolic pathways utilized by alloreactive T cells is fundamental to deepening our understanding of GVHD biology, including its initiation, propagation, and potential mitigation. This review summarizes the metabolic pathways available to alloreactive T cells and highlights key metabolic proteins and pathways linking T cell metabolism to effector function. Our current knowledge of alloreactive T cell metabolism is then explored, showing support for glycolysis, fat oxidation, and glutamine metabolism but also offering a potential explanation for how these presumably contradictory metabolic findings might be reconciled. Examples of additional ways in which metabolism impacts aHSCT are addressed, including the influence of butyrate metabolism on GVHD resolution. Finally, the caveats and challenges of assigning causality using our current metabolic toolbox is discussed, as well as likely future directions in immunometabolism, both to highlight the strengths of the current evidence as well as recognize some of its limitations.
Collapse
Affiliation(s)
- Rebecca A Brown
- Division of Blood and Marrow Transplant and Cellular Therapies, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Craig A Byersdorfer
- Division of Blood and Marrow Transplant and Cellular Therapies, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
14
|
Boucault L, Lopez Robles MD, Thiolat A, Bézie S, Schmueck-Henneresse M, Braudeau C, Vimond N, Freuchet A, Autrusseau E, Charlotte F, Redjoul R, Beckerich F, Leclerc M, Piaggio E, Josien R, Volk HD, Maury S, Cohen JL, Anegon I, Guillonneau C. Transient antibody targeting of CD45RC inhibits the development of graft-versus-host disease. Blood Adv 2020; 4:2501-2515. [PMID: 32511714 PMCID: PMC7284095 DOI: 10.1182/bloodadvances.2020001688] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/24/2020] [Indexed: 02/06/2023] Open
Abstract
Allogeneic bone marrow transplantation (BMT) is a widely spread treatment of many hematological diseases, but its most important side effect is graft-versus-host disease (GVHD). Despite the development of new therapies, acute GVHD (aGVHD) occurs in 30% to 50% of allogeneic BMT and is characterized by the generation of effector T (Teff) cells with production of inflammatory cytokines. We previously demonstrated that a short anti-CD45RC monoclonal antibody (mAb) treatment in a heart allograft rat model transiently decreased CD45RChigh Teff cells and increased regulatory T cell (Treg) number and function allowing long-term donor-specific tolerance. Here, we demonstrated in rat and mouse allogeneic GVHD, as well as in xenogeneic GVHD mediated by human T cells in NSG mice, that both ex vivo depletion of CD45RChigh T cells and in vivo treatment with short-course anti-CD45RC mAbs inhibited aGVHD. In the rat model, we demonstrated that long surviving animals treated with anti-CD45RC mAbs were fully engrafted with donor cells and developed a donor-specific tolerance. Finally, we validated the rejection of a human tumor in NSG mice infused with human cells and treated with anti-CD45RC mAbs. The anti-human CD45RC mAbs showed a favorable safety profile because it did not abolish human memory antiviral immune responses, nor trigger cytokine release in in vitro assays. Altogether, our results show the potential of a prophylactic treatment with anti-human CD45RC mAbs in combination with rapamycin as a new therapy to treat aGVHD without abolishing the antitumor effect.
Collapse
Affiliation(s)
- Laetitia Boucault
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| | - Maria-Dolores Lopez Robles
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| | - Allan Thiolat
- Université Paris-Est Créteil, INSERM, Institut Mondor de Recherche Biomédicale (IMRB), Creteil, France
| | - Séverine Bézie
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| | - Michael Schmueck-Henneresse
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin/Berlin Institute of Health (BIH), Berlin, Germany
| | - Cécile Braudeau
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
- Laboratoire d'Immunologie, Centre d'Immunomonitorage Nantes Atlantique (CIMNA), Centre Hospitalier Universitaire (CHU) Nantes, Nantes, France
| | - Nadège Vimond
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| | - Antoine Freuchet
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| | - Elodie Autrusseau
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| | - Frédéric Charlotte
- Service d'Anatomo-Pathologie, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Rabah Redjoul
- AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Service d'Hematologie Clinique, Creteil, France
| | - Florence Beckerich
- AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Service d'Hematologie Clinique, Creteil, France
| | - Mathieu Leclerc
- AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Service d'Hematologie Clinique, Creteil, France
- Université Paris-Est Créteil, INSERM, IMRB, AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Service d'Hematologie Clinique, Creteil, France
| | - Eliane Piaggio
- Translational Research Department, Institut Curie Research Center, Paris Sciences & Lettres (PSL) Research University, U932, INSERM, Paris, France; and
| | - Regis Josien
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
- Laboratoire d'Immunologie, Centre d'Immunomonitorage Nantes Atlantique (CIMNA), Centre Hospitalier Universitaire (CHU) Nantes, Nantes, France
| | - Hans-Dieter Volk
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin/Berlin Institute of Health (BIH), Berlin, Germany
| | - Sébastien Maury
- Université Paris-Est Créteil, INSERM, IMRB, AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Service d'Hematologie Clinique, Creteil, France
| | - José L Cohen
- Université Paris-Est Créteil, INSERM, IMRB, AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Centre d'Investigation Clinique Biotherapie, Creteil, France
| | - Ignacio Anegon
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| | - Carole Guillonneau
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| |
Collapse
|
15
|
Kim S, Reddy P. Targeting Signal 3 Extracellularly and Intracellularly in Graft-Versus-Host Disease. Front Immunol 2020; 11:722. [PMID: 32411139 PMCID: PMC7198807 DOI: 10.3389/fimmu.2020.00722] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 03/30/2020] [Indexed: 12/19/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HCT) holds curative potential for many hematological disorders. However, the pathophysiology of the desired graft-versus-tumor effect is linked to life-threatening complications of acute graft-versus-host disease (GVHD). Allogeneic donor T lymphocytes are essential for causing GVHD, and their activation relies on the coordination of TCR engagement and co-stimulation, also known as Signal 1 and Signal 2. In addition to these signals, a network of secreted cytokines by immune cells provides a third signal, Signal 3, that is critical for the initiation and maintenance of GVHD. Strategies to target Signal 3 in human diseases have shown therapeutic benefit for inflammatory disorders such as Rheumatoid Arthritis and Inflammatory Bowel Disease. However, despite our growing understanding of their role in GVHD, the success of targeting individual cytokines has been modest with some notable exceptions. This review aims to describe current approaches toward targeting Signal 3 in clinical GVHD, and to highlight emerging studies in immune cell biology that may be harnessed for better clinical translation.
Collapse
Affiliation(s)
- Stephanie Kim
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States.,Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, United States
| | - Pavan Reddy
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
16
|
Considerations for Medications Commonly Utilized in the Oncology Population in the Intensive Care Unit. ONCOLOGIC CRITICAL CARE 2019. [PMCID: PMC7189427 DOI: 10.1007/978-3-319-74588-6_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
An increasing number of oncologic patients are presenting to the intensive care unit with complications from both their chronic disease states and cancer therapies due to improved survival rates. The management of these patients is complex due to immunosuppression (from the malignancy and/or treatment), metabolic complications, and diverse medication regimens with the potential for significant drug-drug interactions and overlapping adverse effects. This chapter will provide clinicians with an overview of non-chemotherapy medications frequently encountered in the critically ill oncologic patient, with a focus on practical considerations.
Collapse
|
17
|
Joo SJ, Yildirim I, Stenger EO, Anderson EJ. Sirolimus-associated pericardial effusion with cardiac tamponade and interstitial pneumonitis in a hematopoietic stem cell transplant recipient. Pediatr Transplant 2019; 23:e13425. [PMID: 31012209 DOI: 10.1111/petr.13425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 02/10/2019] [Accepted: 03/11/2019] [Indexed: 12/24/2022]
Abstract
Sirolimus, a mammalian target of rapamycin (mTOR) inhibitor, is a potent immunosuppressant that is increasingly used in prevention and treatment of graft-vs-host disease (GVHD) in allogeneic hematopoietic stem cell transplant (HSCT) patients. However, data regarding its adverse effects in HSCT patients remain limited. We describe an 18-year-old HSCT patient with a history of invasive fungal infection, who developed pericardial effusion with cardiac tamponade and interstitial pneumonitis while receiving sirolimus for GVHD prophylaxis. Our case illustrates potentially life-threatening complications of sirolimus use in allogeneic HSCT patients.
Collapse
Affiliation(s)
- Su Jin Joo
- Division of Pediatric Infectious Diseases, Nationwide Children's Hospital, Columbus, Ohio.,Division of Pediatric Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Inci Yildirim
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia.,Rollins School of Public Health, Department of Epidemiology, Emory University, Atlanta, Georgia
| | - Elizabeth O Stenger
- Division of Pediatric Hematology and Oncology, Emory University, Atlanta, Georgia
| | - Evan J Anderson
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia.,Division of Infectious Diseases, Department of Internal Medicine, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
18
|
Axt L, Naumann A, Toennies J, Haen SP, Vogel W, Schneidawind D, Wirths S, Moehle R, Faul C, Kanz L, Axt S, Bethge WA. Retrospective single center analysis of outcome, risk factors and therapy in steroid refractory graft-versus-host disease after allogeneic hematopoietic cell transplantation. Bone Marrow Transplant 2019; 54:1805-1814. [PMID: 31089279 DOI: 10.1038/s41409-019-0544-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/24/2019] [Accepted: 04/26/2019] [Indexed: 01/09/2023]
Abstract
Acute and chronic graft-vs.-host disease (aGvHD and cGvHD) are major complications after allogeneic hematopoietic cell transplantation (HCT) leading to substantial morbidity and mortality. This retrospective single-center study analyzes incidence, therapy, and outcome of GvHD in n = 721 patients ≥18 years having received allogeneic HCT 2004-2013 with a special focus on steroid refractory GvHD. Acute (n = 355/49.2%) and chronic (n = 269/37.3%) GvHD were mainly treated by steroids in first-line therapy. The proportion of steroid refractory aGvHD and cGvHD was 35.7% and 31.4%, respectively. As there is no standard therapy for steroid refractory GvHD, a range of different agents was used. In aGvHD, the overall response rate (ORR) of steroid refractory GvHD to second-line treatment was 27.4%. Mycophenolate mofetil (MMF) and mTOR inhibitors led to superior response rates (ORR 50.0% and 53.3%, respectively). In steroid refractory cGvHD therapy, ORR was 44.4%. Use of calcineurin inhibitors (CNI; n = 11/45.5%), MMF (n = 18/50.0%), mTOR inhibitors (n = 10/60.0%), and extracorporeal photophoresis (ECP; n = 16/56.3%) showed ORR above average. Targeted therapies lead to responses in 7.7% (n = 13). This data may help to improve the design of future prospective clinical studies in GvHD.
Collapse
Affiliation(s)
- L Axt
- Department of Hematology and Oncology, Medical Center University Hospital Tuebingen, Tuebingen, Germany
| | - A Naumann
- Institute for Clinical Epidemiology and Applied Biometry, Eberhard-Karl University Tuebingen, Tuebingen, Germany
| | - J Toennies
- Department of Hematology and Oncology, Medical Center University Hospital Tuebingen, Tuebingen, Germany
| | - S P Haen
- Department of Hematology and Oncology, Medical Center University Hospital Tuebingen, Tuebingen, Germany
| | - W Vogel
- Department of Hematology and Oncology, Medical Center University Hospital Tuebingen, Tuebingen, Germany
| | - D Schneidawind
- Department of Hematology and Oncology, Medical Center University Hospital Tuebingen, Tuebingen, Germany
| | - S Wirths
- Department of Hematology and Oncology, Medical Center University Hospital Tuebingen, Tuebingen, Germany
| | - R Moehle
- Department of Hematology and Oncology, Medical Center University Hospital Tuebingen, Tuebingen, Germany
| | - C Faul
- Department of Hematology and Oncology, Medical Center University Hospital Tuebingen, Tuebingen, Germany
| | - L Kanz
- Department of Hematology and Oncology, Medical Center University Hospital Tuebingen, Tuebingen, Germany
| | - S Axt
- Department for Visceral, General and Transplant Surgery, University Hospital Tuebingen, Tuebingen, Germany
| | - W A Bethge
- Department of Hematology and Oncology, Medical Center University Hospital Tuebingen, Tuebingen, Germany.
| |
Collapse
|
19
|
Griesenauer B, Jiang H, Yang J, Zhang J, Ramadan AM, Egbosiuba J, Campa K, Paczesny S. ST2/MyD88 Deficiency Protects Mice against Acute Graft-versus-Host Disease and Spares Regulatory T Cells. THE JOURNAL OF IMMUNOLOGY 2019; 202:3053-3064. [PMID: 30979817 DOI: 10.4049/jimmunol.1800447] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 03/18/2019] [Indexed: 12/11/2022]
Abstract
Acute graft-versus-host disease (aGVHD) hinders the efficacy of allogeneic hematopoietic cell transplantation (HCT). Plasma levels of soluble membrane-bound ST2 (ST2) are elevated in human and murine aGVHD and correlated to type 1 T cells response. ST2 signals through the adapter protein MyD88. The role of MyD88 in T cells during aGVHD has yet to be elucidated. We found that knocking out MyD88 in the donor T cells protected against aGVHD independent of IL-1R and TLR4 signaling in two murine HCT models. This protection was entirely driven by MyD88-/- CD4 T cells. Transplanting donor MyD88-/- conventional T cells (Tcons) with wild-type (WT) or MyD88-/- regulatory T cells (Tregs) lowered aGVHD severity and mortality. Transcriptome analysis of sorted MyD88-/- CD4 T cells from the intestine 10 d post-HCT showed lower levels of Il1rl1 (gene of ST2), Ifng, Csf2, Stat5, Batf, and Jak2 Transplanting donor ST2-/- Tcons with WT or ST2-/- Tregs showed a similar phenotype with what we observed when using donor MyD88-/- Tcons. Decreased ST2 was confirmed at the protein level with less secretion of soluble ST2 and more expression of ST2 compared with WT T cells. Our data suggest that Treg suppression from lack of MyD88 signaling in donor Tcons during alloreactivity uses the ST2 but not the IL-1R or TLR4 pathways, and ST2 represents a potential aGVHD therapeutic target sparing Tregs.
Collapse
Affiliation(s)
| | - Hua Jiang
- Indiana University School of Medicine, Indianapolis, IN 46202
| | - Jinfeng Yang
- Indiana University School of Medicine, Indianapolis, IN 46202
| | - Jilu Zhang
- Indiana University School of Medicine, Indianapolis, IN 46202
| | | | - Jane Egbosiuba
- Indiana University School of Medicine, Indianapolis, IN 46202
| | - Khaled Campa
- Indiana University School of Medicine, Indianapolis, IN 46202
| | - Sophie Paczesny
- Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
20
|
FL/GCSF/AMD3100-mobilized Hematopoietic Stem Cells Induce Mixed Chimerism With Nonmyeloablative Conditioning and Transplantation Tolerance. Transplantation 2019; 103:1360-1371. [PMID: 30747856 DOI: 10.1097/tp.0000000000002657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Mobilization of hematopoietic stem cells (HSCs) has become the preferred approach for HSC transplantation. AMD3100, a competitive inhibitor of C-X-C motif chemokine receptor-4, has been found to be a rapid mobilizing agent. The present study evaluated approaches to optimize the product collected. METHODS Mobilized peripheral blood mononuclear cells (mPBMCs) from B6 mice were transplanted to recipient BALB/c mice conditioned with ablative or nonmyeloablative approaches. RESULTS The optimal dose of AMD3100 was found to be 5.0 mg/kg. Optimal HSC mobilization was observed when AMD3100 (day 10) was coadministered with Flt3 ligand (FL) (days 1-10) and granulocyte colony-stimulating factor (GCSF) (days 4-10). There was a 228.8-fold increase of HSC with FL/GCSF/AMD3100 compared with AMD3100 treatment alone. When unmodified mPBMCs were transplanted into ablated allogeneic recipients, all recipients expired by day 40 from severe acute graft versus host disease (GVHD). When T cells were depleted from mPBMC, long-term survival and engraftment were achieved in majority of the recipients. When PBMC mobilized by FL/GCSF/AMD3100 were transplanted into recipients conditioned nonmyeloablatively with anti-CD154/rapamycin plus 100, 200, and 300 cGy of total body irradiation, 42.9%, 85.7%, and 100% of mice engrafted, respectively. Donor chimerism was durable, multilineage, and stable. Lymphocytes from mixed chimeras showed no response to host or donor antigens, suggesting functional bidirection T-cell tolerance in vitro. Most importantly, none of the engrafted mice exhibited clinical features of GVHD. CONCLUSIONS FL/GCSF/AMD3100 is an efficient treatment to maximally mobilize HSC. Durable engraftment and donor-specific tolerance can be achieved with mPBMC in nonmyeloablative conditioning without GVHD.
Collapse
|
21
|
Cross Talk Networks of Mammalian Target of Rapamycin Signaling With the Ubiquitin Proteasome System and Their Clinical Implications in Multiple Myeloma. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 343:219-297. [PMID: 30712673 DOI: 10.1016/bs.ircmb.2018.06.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Multiple myeloma (MM) is the second most common hematological malignancy and results from the clonal amplification of plasma cells. Despite recent advances in treatment, MM remains incurable with a median survival time of only 5-6years, thus necessitating further insights into MM biology and exploitation of novel therapeutic approaches. Both the ubiquitin proteasome system (UPS) and the PI3K/Akt/mTOR signaling pathways have been implicated in the pathogenesis, and treatment of MM and different lines of evidence suggest a close cross talk between these central cell-regulatory signaling networks. In this review, we outline the interplay between the UPS and mTOR pathways and discuss their implications for the pathophysiology and therapy of MM.
Collapse
|
22
|
Lin Y, Wang B, Shan W, Tan Y, Feng J, Xu L, Wang L, Han B, Zhang M, Yu J, Yu X, Huang H. mTOR inhibitor rapamycin induce polymorphonuclear myeloid-derived suppressor cells mobilization and function in protecting against acute graft-versus-host disease after bone marrow transplantation. Clin Immunol 2017; 187:122-131. [PMID: 29132870 DOI: 10.1016/j.clim.2017.11.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 10/23/2017] [Accepted: 11/09/2017] [Indexed: 01/07/2023]
Abstract
The mammalian target of rapamycin (mTOR) inhibitor rapamycin (RAPA) has been shown to be an effective immunosuppressor in the management of acute graft-versus-host disease (aGVHD) after bone marrow transplantation. Myeloid-derived suppressor cells (MDSCs) also have a protective effect in aGVHD regulation. However, the relationship between RAPA and MDSCs in aGVHD models is unclear. Meanwhile, the effect of RAPA on different subgroups of MDSCs is also less well described. In this study, we demonstrate that in vivo administration of RAPA results in the expansion and functional enhancement of polymorphonuclear MDSCs (PMN-MDSCs) in a murine model of aGVHD. RAPA treatment can enhance the suppressive function of PMN-MDSCs via up-regulation of arginase1 (Arg1) and induced nitric oxide synthase (iNOS) at later time points. Moreover, RAPA can also induce a strong immunosuppressive function in PMN-MDSCs from murine bone marrow in vitro, but has a contrary effect on monocytic MDSCs (M-MDSCs). We found that RAPA-treated PMN-MDSCs can restrain the differentiation of Th1/Th2 cells and promote induction of regulatory T cells in in vitro studies.
Collapse
Affiliation(s)
- Yu Lin
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, China
| | - Binsheng Wang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wei Shan
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yamin Tan
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jingjing Feng
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lin Xu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Limengmeng Wang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, China
| | - Biqing Han
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Mingming Zhang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jian Yu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaohong Yu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, China.
| |
Collapse
|
23
|
Chao YH, Chang YC, Wu HP, Peng CT, Weng TF, Wu KH. Everolimus for pediatric patients with acute graft-versus-host disease after hematopoietic stem cell transplantation: A pilot study. Medicine (Baltimore) 2017; 96:e8464. [PMID: 29095297 PMCID: PMC5682816 DOI: 10.1097/md.0000000000008464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Acute graft-versus-host disease (aGVHD) is a significant cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (HSCT). Due to the poor prognosis for patients not responding to first-line steroids treatment, improvements in aGVHD therapy are needed. Everolimus is a promising candidate that combines immunosuppressive properties with anti-neoplastic effects. Here, we retrospectively reviewed the efficacy of everolimus with steroids as primary treatment in 13 patients with grade II to grade IV aGVHD after HSCT. Among them, 12 (92.3%) had complete response to everolimus with steroids without additional immunosuppressive agents. The median duration of therapy was 76 days (range 20-110). Asymptomatic hypertriglyceridemia was the most common therapy complication (69.2%), but treatment interruption was not needed. Thrombotic microangiopathy was rare (7.7%), but can be quickly solved by stopping everolimus and cyclosporine treatment. Other toxicities were manageable. Two patients developed chronic GVHD (15.4%), limited in one and extensive in the other. The overall survival was 76.9% with a median follow-up of 3.4 years after HSCT (range 0.7-5.7). Accordingly, everolimus with steroids were feasible for patients with aGVHD after HSCT as primary treatment. Further large-scale studies are required.
Collapse
Affiliation(s)
- Yu-Hua Chao
- Department of Pediatrics, Chung Shan Medical University Hospital
- School of Medicine, Chung Shan Medical University
| | - Yin-Chen Chang
- Department of Nursing, College of Medicine and Nursing, Hungkuang University
| | - Han-Ping Wu
- Division of Pediatric General Medicine, Department of Pediatrics, Chang Gung Memorial Hospital
- College of Medicine, Chang Gung University, Taoyuan
| | - Ching-Tien Peng
- Division of Pediatric Hematology-Oncology, Children's Hospital, China Medical University
- Department of Biotechnology and Bioinformatics, Asia University
| | - Te-Fu Weng
- Division of Pediatric Hematology-Oncology, Children's Hospital, China Medical University
| | - Kang-Hsi Wu
- Division of Pediatric Hematology-Oncology, Children's Hospital, China Medical University
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
24
|
Lutz M, Mielke S. New perspectives on the use of mTOR inhibitors in allogeneic haematopoietic stem cell transplantation and graft-versus-host disease. Br J Clin Pharmacol 2016; 82:1171-1179. [PMID: 27245261 PMCID: PMC5061796 DOI: 10.1111/bcp.13022] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/27/2016] [Accepted: 05/30/2016] [Indexed: 12/17/2022] Open
Abstract
Inhibition of the mechanistic target of rapamycin (mTOR) has been exploited largely both in solid tumour oncology and solid organ transplantation. More recently mTOR inhibitors such as sirolimus and everolimus have been introduced to the field of allogeneic haematopoietic stem cell transplantation where their unique combination of immunosuppressive purposes offering reduced nephrotoxicity and potential antimalignant effects reflect a unique drug profile that has led to their widespread use in both prophylaxis and therapy of graft-versus-host disease (GVHD). On the other hand haematological insufficiency, infectious complications as well as vasculopathies, have been frequently reported as limiting toxicities. Here, we review both the retrospective and prospective experience available to date and stress the need for prospective registration trials to reduce off label use and improve patient safety by optimizing dosing and enhancing pharmacovigilance. Furthermore, we speculate on the future role of mTOR inhibitors in allogeneic haematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Mathias Lutz
- Department of Medicine A, Münster University Medical Center, Münster, Germany
| | - Stephan Mielke
- Department of Internal Medicine II, Würzburg University Medical Center, Würzburg, Germany.
| |
Collapse
|
25
|
Herrero-Sánchez MC, Rodríguez-Serrano C, Almeida J, San Segundo L, Inogés S, Santos-Briz Á, García-Briñón J, Corchete LA, San Miguel JF, Del Cañizo C, Blanco B. Targeting of PI3K/AKT/mTOR pathway to inhibit T cell activation and prevent graft-versus-host disease development. J Hematol Oncol 2016; 9:113. [PMID: 27765055 PMCID: PMC5072323 DOI: 10.1186/s13045-016-0343-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 10/08/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Graft-versus-host disease (GvHD) remains the major obstacle to successful allogeneic hematopoietic stem cell transplantation, despite of the immunosuppressive regimens administered to control T cell alloreactivity. PI3K/AKT/mTOR pathway is crucial in T cell activation and function and, therefore, represents an attractive therapeutic target to prevent GvHD development. Recently, numerous PI3K inhibitors have been developed for cancer therapy. However, few studies have explored their immunosuppressive effect. METHODS The effects of a selective PI3K inhibitor (BKM120) and a dual PI3K/mTOR inhibitor (BEZ235) on human T cell proliferation, expression of activation-related molecules, and phosphorylation of PI3K/AKT/mTOR pathway proteins were analyzed. Besides, the ability of BEZ235 to prevent GvHD development in mice was evaluated. RESULTS Simultaneous inhibition of PI3K and mTOR was efficient at lower concentrations than PI3K specific targeting. Importantly, BEZ235 prevented naïve T cell activation and induced tolerance of alloreactive T cells, while maintaining an adequate response against cytomegalovirus, more efficiently than BKM120. Finally, BEZ235 treatment significantly improved the survival and decreased the GvHD development in mice. CONCLUSIONS These results support the use of PI3K inhibitors to control T cell responses and show the potential utility of the dual PI3K/mTOR inhibitor BEZ235 in GvHD prophylaxis.
Collapse
Affiliation(s)
- Mª Carmen Herrero-Sánchez
- Servicio de Hematología, Hospital Universitario de Salamanca, Paseo de San Vicente 58-182, 37007, Salamanca, Spain.,Instituto de Investigación Biomédica de Salamanca (IBSAL), Paseo de San Vicente 58-182, 37007, Salamanca, Spain.,Centro de Investigación del Cáncer, Universidad de Salamanca, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Concepción Rodríguez-Serrano
- Servicio de Hematología, Hospital Universitario de Salamanca, Paseo de San Vicente 58-182, 37007, Salamanca, Spain.,Instituto de Investigación Biomédica de Salamanca (IBSAL), Paseo de San Vicente 58-182, 37007, Salamanca, Spain.,Centro de Investigación del Cáncer, Universidad de Salamanca, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Julia Almeida
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Paseo de San Vicente 58-182, 37007, Salamanca, Spain.,Centro de Investigación del Cáncer, Universidad de Salamanca, Campus Miguel de Unamuno, 37007, Salamanca, Spain.,Servicio de Citometría, Centro de Investigación del Cáncer, Universidad de Salamanca, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Laura San Segundo
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Paseo de San Vicente 58-182, 37007, Salamanca, Spain.,Centro de Investigación del Cáncer, Universidad de Salamanca, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Susana Inogés
- Laboratorio de Inmunoterapia, Clínica Universidad de Navarra, Avda. Pío XII 55, 31008, Pamplona, Spain
| | - Ángel Santos-Briz
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Paseo de San Vicente 58-182, 37007, Salamanca, Spain.,Departamento de Patología, Hospital Universitario de Salamanca, Paseo de San Vicente 58-182, 37007, Salamanca, Spain
| | - Jesús García-Briñón
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Paseo de San Vicente 58-182, 37007, Salamanca, Spain.,Departamento de Biología Celular y Patología, Facultad de Medicina, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Luis Antonio Corchete
- Servicio de Hematología, Hospital Universitario de Salamanca, Paseo de San Vicente 58-182, 37007, Salamanca, Spain.,Instituto de Investigación Biomédica de Salamanca (IBSAL), Paseo de San Vicente 58-182, 37007, Salamanca, Spain.,Centro de Investigación del Cáncer, Universidad de Salamanca, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Jesús F San Miguel
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra, Avda. Pío XII 55, 31008, Pamplona, Spain
| | - Consuelo Del Cañizo
- Servicio de Hematología, Hospital Universitario de Salamanca, Paseo de San Vicente 58-182, 37007, Salamanca, Spain.,Instituto de Investigación Biomédica de Salamanca (IBSAL), Paseo de San Vicente 58-182, 37007, Salamanca, Spain.,Centro de Investigación del Cáncer, Universidad de Salamanca, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Belén Blanco
- Servicio de Hematología, Hospital Universitario de Salamanca, Paseo de San Vicente 58-182, 37007, Salamanca, Spain. .,Instituto de Investigación Biomédica de Salamanca (IBSAL), Paseo de San Vicente 58-182, 37007, Salamanca, Spain. .,Centro de Investigación del Cáncer, Universidad de Salamanca, Campus Miguel de Unamuno, 37007, Salamanca, Spain.
| |
Collapse
|
26
|
Herrero-Sánchez MC, Rodríguez-Serrano C, Almeida J, San-Segundo L, Inogés S, Santos-Briz Á, García-Briñón J, SanMiguel JF, Del Cañizo C, Blanco B. Effect of mTORC1/mTORC2 inhibition on T cell function: potential role in graft-versus-host disease control. Br J Haematol 2016; 173:754-68. [PMID: 26914848 DOI: 10.1111/bjh.13984] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 12/21/2015] [Indexed: 12/17/2022]
Abstract
The mechanistic target of rapamycin (mTOR) pathway is crucial for the activation and function of T cells, which play an essential role in the development of graft-versus-host disease (GvHD). Despite its partial ability to block mTOR pathway, the mTORC1 inhibitor rapamycin has shown encouraging results in the control of GvHD. Therefore, we considered that simultaneous targeting of both mTORC1 and mTORC2 complexes could exert a more potent inhibition of T cell activation and, thus, could have utility in GvHD control. To assess this assumption, we have used the dual mTORC1/mTORC2 inhibitors CC214-1 and CC214-2. In vitro studies confirmed the superior ability of CC214-1 versus rapamycin to block mTORC1 and mTORC2 activity and to reduce T cell proliferation. Both drugs induced a similar decrease in Th1/Th2 cytokine secretion, but CC214-1 was more efficient in inhibiting naïve T cell activation and the expression of T-cell activation markers. In addition, CC214-1 induced specific tolerance against alloantigens, while preserving anti-cytomegalovirus response. Finally, in a mouse model of GvHD, the administration of CC214-2 significantly improved mice survival and decreased GvHD-induced damages. In conclusion, the current study shows, for the first time, the immunosuppressive ability of CC214-1 on T lymphocytes and illustrates the role of CC214-2 in the allogeneic transplantation setting as a possible GvHD prophylaxis agent.
Collapse
Affiliation(s)
- Ma Carmen Herrero-Sánchez
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain.,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.,Centro de Investigación del Cáncer, Universidad de Salamanca, Salamanca, Spain
| | - Concepción Rodríguez-Serrano
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain.,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.,Centro de Investigación del Cáncer, Universidad de Salamanca, Salamanca, Spain
| | - Julia Almeida
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.,Servicio de Citometría, Centro de Investigación del Cáncer, Universidad de Salamanca, Salamanca, Spain
| | - Laura San-Segundo
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.,Centro de Investigación del Cáncer, Universidad de Salamanca, Salamanca, Spain
| | - Susana Inogés
- Laboratorio de Inmunoterapia, Clínica Universidad de Navarra, Pamplona, Spain
| | - Ángel Santos-Briz
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.,Departamento de Patología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Jesús García-Briñón
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.,Departamento de Biología Celular y Patología, Facultad de Medicina, Salamanca, Spain
| | - Jesús F SanMiguel
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Consuelo Del Cañizo
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain.,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.,Centro de Investigación del Cáncer, Universidad de Salamanca, Salamanca, Spain
| | - Belén Blanco
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain.,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| |
Collapse
|
27
|
Abstract
Diabetes mellitus is a metabolic homeostasis disease that contributes to additional comorbidities such as cardiovascular disease (CVD) and cancer. It has a long undiagnosed latent period during which there can be irreparable damage to the pancreas and cardiovascular tissues. Recent studies have highlighted the roles of several microRNAs in CVD. Determining the microRNAs that link diabetes mellitus and CVD is an important topic to be explored. In the present review, we discuss the microRNAs that contribute to the progression of diabetes mellitus and CVD and focus on the miR-29 family microRNAs whose expression is upregulated by hyperglycemia and proinflammatory cytokines, the hallmarks of diabetes mellitus. Upregulation of miR-29 expression is a key factor in the loss of pancreatic β cells and development of the first stage of type 1 diabetes mellitus (T1DM). Additionally, miR-29-mediated suppression of myeloid cell leukemia 1 (MCL-1), an important prosurvival protein, underlies Marfan's syndrome, abdominal aortic aneurysm, and diabetes mellitus-associated cardiomyocyte disorganization. Suppression of miR-29 expression and subsequent increase in the prosurvival MCL-1, however, promotes tumor development. Therefore, miR-29 mimics that suppress MCL-1 are hailed as tumor suppressors. The critical question is whether an increase in miR-29 levels is well tolerated in conditions of comorbidities in which insulin resistance is an underlying disease. In light of increasing awareness of the interconnection of diabetes mellitus, CVD, and cancer, it is of utmost importance to understand the mechanism of action of current treatment options on all of the comorbidities and careful evaluation of cardiovascular toxicity must accompany any treatment paradigm that increases miR-29 levels.
Collapse
Affiliation(s)
- Anna Ślusarz
- aDepartment of Medicine bDepartment of Biochemistry, University of Missouri cHarry S. Truman Memorial Veterans Affairs Hospital dDepartment of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, USA
| | | |
Collapse
|
28
|
Weber T, Niestadtkötter J, Wienke A, Müller-Tidow C, Müller LP. Enteric-coated mycophenolate sodium containing GvHD prophylaxis reduces GvHD rate after allogeneic HSCT. Eur J Haematol 2016; 97:232-8. [PMID: 26613546 DOI: 10.1111/ejh.12710] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2015] [Indexed: 12/24/2022]
Abstract
OBJECTIVES The aim of this study was to evaluate whether cyclosporine A (CsA)-based Graft vs. Host Disease (GvHD) prophylaxis with enteric-coated mycophenolate sodium (EC-MPS) instead of mycophenolate mofetil (MMF) or methotrexate (MTX) reduces the GvHD incidence and lowers gastrointestinal (GI-) toxicities. METHODS In a retrospective analysis of 102 allogeneic hematopoietic stem cell transplant (HSCT) patients, incidences of overall and severe aGvHD (>II°), cGvHD as well as overall and severe (CTC >II°) GI-toxicities were compared between GvHD prophylaxis containing EC-MPS vs. MMF or MTX (control group). RESULTS The overall aGvHD rate was significantly lower in the EC-MPS group compared to the control (47% vs. 72%, P = 0.022) with lower rates of severe aGvHD (10% vs. 25%, P = 0.088) and cGvHD (20% vs. 39%, P = 0.065). Prophylaxis with EC-MPS remained significantly associated with a lower aGvHD rate in a multiple logistic regression model. GI-toxicities did not differ between both groups except for severe abdominal pain for which the incidence was increased in the EC-MPS group (17% vs. 3%, P = 0.022). CONCLUSIONS This data support the hypothesis that replacement of MMF or MTX by EC-MPS reduces GvHD rates after HSCT. This appears not to be due to a reduced GI-toxicity of EC-MPS.
Collapse
Affiliation(s)
| | | | - Andreas Wienke
- Institute of Medical Epidemiology, Biostatistics and Informatics, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | | | | |
Collapse
|
29
|
García E, Buenasmañanas D, Martín C, Rojas R. [Sirolimus associated pneumonitis in a hematopoietic stem cell transplant patient]. Med Clin (Barc) 2015; 145:21-3. [PMID: 25978917 DOI: 10.1016/j.medcli.2015.02.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 02/14/2015] [Accepted: 02/26/2015] [Indexed: 11/27/2022]
Abstract
BACKGROUND AND OBJECTIVE Sirolimus (SR) is a lipophilic macrocytic lactone with immunosuppressive properties (mTOR inhibitor) commonly used in solid organ transplantation and recently introduced in the prophylaxis and treatment of graft-versus-host disease. Its numerous side effects include: hyperlipidemia, arthralgias, noncardiac peripheral edema, thrombotic microangiopathy and interstitial pneumonitis. SR-associated pneumonitis is a rare but potentially serious complication due to its increasing utilization in transplant patients. PATIENT AND METHOD We report the case of a patient undergoing hematopoietic stem cell transplantation with severe respiratory distress and SR therapy. RESULTS Microbiological tests were all negative and other complications related to transplantation were discarded. The chest computed tomography of high-resolution showed pneumonitis. The SR therapy was interrupted and treatment was started with steroids with resolution of symptoms. CONCLUSIONS SR associated pneumonitis is a potentially fatal side effect. In patients treated with SR and respiratory failure, we must suspect this complication because early recognition along with drug discontinuation and steroid treatment is essential to reverse this complication.
Collapse
Affiliation(s)
- Estefanía García
- Servicio de Hematología, Hospital Universitario Reina Sofía, Córdoba, España
| | - Diana Buenasmañanas
- Servicio de Hematología, Hospital Universitario Reina Sofía, Córdoba, España
| | - Carmen Martín
- Servicio de Hematología, Hospital Universitario Reina Sofía, Córdoba, España
| | - Rafael Rojas
- Servicio de Hematología, Hospital Universitario Reina Sofía, Córdoba, España.
| |
Collapse
|
30
|
Shook DR, Triplett BM, Eldridge PW, Kang G, Srinivasan A, Leung W. Haploidentical stem cell transplantation augmented by CD45RA negative lymphocytes provides rapid engraftment and excellent tolerability. Pediatr Blood Cancer 2015; 62:666-73. [PMID: 25559618 DOI: 10.1002/pbc.25352] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 10/20/2014] [Indexed: 12/31/2022]
Abstract
BACKGROUND Haploidentical donors are being increasingly used for allogeneic hematopoietic cell transplantation (HCT). However, the requisite T-cell depletion results in a profound and often long-lasting immunocompromised state, and donor lymphocyte infusions bring a risk of graft-versus-host disease (GVHD). Naïve T-cells are believed to be among the most alloreactive T-cell subset and can be identified by CD45RA expression. Allogeneic HCT using CD45RA depletion has not been previously described for haploidentical donors. PROCEDURE Eight children with relapsed or refractory solid tumors were transplanted following myeloablative conditioning. Each patient received two cell products, one created by CD3 depletion and the other through CD45RA depletion. RESULTS Median CD34 recovery was 59.2% with CD45RA depletion, compared to 82.4% using CD3 depletion. Median CD3+ T-cell dose after CD45RA reduction was 99.2 × 10(6) cells/kg, yet depletion of CD3+ CD45RA+ cells exceeded 4.5 log. CD45RA depletion also resulted in substantial depletion of B-cells (median 2.45 log). All eight patients engrafted within 14 days and rapidly achieved 100% donor chimerism. No acute GVHD or secondary graft failure was observed. CONCLUSIONS CD45RA depletion is a novel approach to haploidentical HCT that offers rapid engraftment with minimal risk of GVHD.
Collapse
Affiliation(s)
- David R Shook
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee 38105; Department of Pediatrics, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee 38163
| | | | | | | | | | | |
Collapse
|
31
|
[Delayed wound healing during therapy of cutaneous graft-versus-host disease with everolimus]. Hautarzt 2015; 65:553-5. [PMID: 24577262 DOI: 10.1007/s00105-014-2762-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Graft-versus-host-disease (GvHD) is despite improvement in transplantation medicine the major cause for morbidity and mortality after allogeneic stem cell transplantation. We describe a patient with chronic cutaneous GvHD who developed massive skin ulcerations after changing the immunosuppressive therapy to a mammalian target of rapamycin (mTOR)-inhibitor.
Collapse
|
32
|
Villa A, Aboalela A, Luskin KA, Cutler CS, Sonis ST, Woo SB, Peterson DE, Treister NS. Mammalian target of rapamycin inhibitor-associated stomatitis in hematopoietic stem cell transplantation patients receiving sirolimus prophylaxis for graft-versus-host disease. Biol Blood Marrow Transplant 2014; 21:503-8. [PMID: 25482865 DOI: 10.1016/j.bbmt.2014.11.680] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 11/24/2014] [Indexed: 10/24/2022]
Abstract
The mammalian target of rapamycin (mTOR) inhibitor sirolimus is effective in reducing incidence of graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (HSCT). Agents that inhibit the mTOR pathway are known to be associated with significant and potentially dose-limiting toxicities, including stomatitis. The objective of this study was to report the clinical features and management outcomes of sirolimus-associated oral ulcers in the context of post-HSCT prophylaxis of GVHD. Seventeen patients, from a study cohort of 967, who were treated with sirolimus as prophylaxis for GVHD after allogeneic HSCT at the Dana-Farber/Brigham and Women's Cancer Center developed oral ulcers and were referred to the oral medicine clinic for evaluation and treatment over a period of 6 years. Clinical characteristics (appearance, anatomic site, size) and therapeutic outcomes (time to complete resolution) were documented. Median time to onset of oral ulceration was 55 days after allogeneic HSCT (range, 6 to 387 days); 92.9% of ulcers were located on nonkeratinized mucosa, with the ventrolateral tongue the most common site of involvement. Thirteen patients were treated with topical corticosteroid therapy; 12 of these patients also required intralesional corticosteroid injections. Clinical improvement (resolution of the lesions and improvement of symptoms) was noted in all cases, with no reported adverse events. Median time to complete resolution after onset of therapy was 14 days (range, 2 to 70 days). Patients receiving sirolimus for GVHD prophylaxis may develop painful oral ulcerations, which can be effectively managed with topical steroid treatment. Further prospective studies are needed to better elucidate the incidence of this complication, identify risk factors, and evaluate the effectiveness of interventions.
Collapse
Affiliation(s)
- Alessandro Villa
- Division of Oral Medicine and Dentistry, Brigham and Women's Hospital, and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts.
| | - Ali Aboalela
- Division of Oral Medicine and Dentistry, Brigham and Women's Hospital, and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Katharine A Luskin
- School of Dental Medicine, University of Connecticut Health Center, Farmington, Connecticut
| | - Corey S Cutler
- Department of Medical Oncology, Brigham & Women's Hospital/Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Stephen T Sonis
- Division of Oral Medicine and Dentistry, Brigham and Women's Hospital, and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Sook Bin Woo
- Division of Oral Medicine and Dentistry, Brigham and Women's Hospital, and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Douglas E Peterson
- School of Dental Medicine, University of Connecticut Health Center, Farmington, Connecticut; Department of Oral Health and Diagnostic Sciences, School of Dental Medicine and Neag Comprehensive Cancer Center, University of Connecticut Health Center, Farmington, Connecticut
| | - Nathaniel S Treister
- Division of Oral Medicine and Dentistry, Brigham and Women's Hospital, and Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts
| |
Collapse
|
33
|
Mutalib M, Borrelli O, Blackstock S, Kiparissi F, Elawad M, Shah N, Lindley K. The use of sirolimus (rapamycin) in the management of refractory inflammatory bowel disease in children. J Crohns Colitis 2014; 8:1730-4. [PMID: 25240477 DOI: 10.1016/j.crohns.2014.08.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 08/24/2014] [Accepted: 08/31/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND Management of refractory inflammatory bowel disease (IBD) in children is challenging and once response to conventional medical therapy deviates from the expected, options are often limited. Sirolimus is commonly used in post-transplantation management and is used sparsely as rescue therapy in refractory Crohn's disease. In the present study, we report the efficacy of sirolimus as an adjuvant immunosuppressive therapy in a retrospective case review of a selected group of IBD children who were refractory to the conventional treatments. METHODS Medical records of children with refractory IBD unresponsive to conventional therapy and started on sirolimus between 2006 and 2012 were retrospectively reviewed. Clinical response, through Pediatric Ulcerative Colitis Activity Index (PUCAI) and Pediatric Crohn's Disease Activity Index (PCDAI), as well as intestinal inflammation, through specific histological scores, was evaluated. RESULTS The records of 14 patients were analyzed. Eleven of them had ulcerative colitis (UC) and 3 Crohn's disease (CD); mean age at diagnosis was 9.1 years (standard deviation 3.8). Of UC patients, 5 (45%) achieved clinical remission and 2 (18%) showed clinical response. All CD patients went into clinical remission. Mucosal healing was achieved by 5 children (45%) with UC and 2 (67%) with CD patients. One child with ulcerative colitis was weaned off adalimumab, while 2 children with CD were weaned off prednisolone and methotrexate successfully. CONCLUSION Our data provide evidence that sirolimus seems to be effective as rescue therapy in a subgroup of children with severe IBD refractory to conventional therapies by inducing both clinical remission and mucosal healing.
Collapse
Affiliation(s)
- Mohamed Mutalib
- Department of Paediatric Gastroenterology, Great Ormond Street Hospital for Sick Children NHS Foundation Trust, United Kingdom.
| | - Osvaldo Borrelli
- Department of Paediatric Gastroenterology, Great Ormond Street Hospital for Sick Children NHS Foundation Trust, United Kingdom
| | - Sarah Blackstock
- Department of Paediatric Gastroenterology, Great Ormond Street Hospital for Sick Children NHS Foundation Trust, United Kingdom
| | - Fevronia Kiparissi
- Department of Paediatric Gastroenterology, Great Ormond Street Hospital for Sick Children NHS Foundation Trust, United Kingdom
| | - Mamoun Elawad
- Department of Paediatric Gastroenterology, Great Ormond Street Hospital for Sick Children NHS Foundation Trust, United Kingdom
| | - Neil Shah
- Department of Paediatric Gastroenterology, Great Ormond Street Hospital for Sick Children NHS Foundation Trust, United Kingdom
| | - Keith Lindley
- Department of Paediatric Gastroenterology, Great Ormond Street Hospital for Sick Children NHS Foundation Trust, United Kingdom
| |
Collapse
|
34
|
Valizadeh H, Ghanbarzadeh S, Zakeri-Milani P. Fusogenic liposomal formulation of sirolimus: improvement of drug anti-proliferative effect on human T-cells. Drug Dev Ind Pharm 2014; 41:1558-65. [DOI: 10.3109/03639045.2014.971032] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
35
|
Li L, Torres-Coronado M, Gu A, Rao A, Gardner AM, Epps EW, Gonzalez N, Tran CA, Wu X, Wang JH, DiGiusto DL. Enhanced genetic modification of adult growth factor mobilized peripheral blood hematopoietic stem and progenitor cells with rapamycin. Stem Cells Transl Med 2014; 3:1199-208. [PMID: 25107584 DOI: 10.5966/sctm.2014-0010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Genetic modification of adult human hematopoietic stem and progenitor cells (HSPCs) with lentiviral vectors leads to long-term gene expression in the progeny of the HSPCs and has been used to successfully treat several monogenic diseases. In some cases, the gene-modified cells have a selective growth advantage over nonmodified cells and eventually are the dominant engrafted population. However, in disease indications for which the gene-modified cells do not have a selective advantage, optimizing transduction of HSPC is paramount to successful stem cell-based gene therapy. We demonstrate here that transduction of adult CD34+ HSPCs with lentiviral vectors in the presence of rapamycin, a widely used mTORC1 inhibitor, results in an approximately threefold increase in stable gene marking with minimal effects on HSPC growth and differentiation. Using this approach, we have demonstrated that we can enhance the frequency of gene-modified HSPCs that give rise to clonogenic progeny in vitro without excessive increases in the number of vector copies per cell or changes in integration pattern. The genetic marking of HSPCs and expression of transgenes is durable, and transplantation of gene-modified HSPCs into immunodeficient mice results in high levels of gene marking of the lymphoid and myeloid progeny in vivo. The prior safe clinical history of rapamycin in other applications supports the use of this compound to generate gene-modified autologous HSPCs for our HIV gene therapy clinical trials.
Collapse
Affiliation(s)
- Lijing Li
- Shared Resources-Cellular Process, City of Hope, Duarte, California, USA; Center for Blood Cell Therapies at Peter McCallum Center, Melbourne, Australia; Departments of Molecular and Cellular Biology and Virology, City of Hope, Duarte, California, USA
| | - Mónica Torres-Coronado
- Shared Resources-Cellular Process, City of Hope, Duarte, California, USA; Center for Blood Cell Therapies at Peter McCallum Center, Melbourne, Australia; Departments of Molecular and Cellular Biology and Virology, City of Hope, Duarte, California, USA
| | - Angel Gu
- Shared Resources-Cellular Process, City of Hope, Duarte, California, USA; Center for Blood Cell Therapies at Peter McCallum Center, Melbourne, Australia; Departments of Molecular and Cellular Biology and Virology, City of Hope, Duarte, California, USA
| | - Anitha Rao
- Shared Resources-Cellular Process, City of Hope, Duarte, California, USA; Center for Blood Cell Therapies at Peter McCallum Center, Melbourne, Australia; Departments of Molecular and Cellular Biology and Virology, City of Hope, Duarte, California, USA
| | - Agnes M Gardner
- Shared Resources-Cellular Process, City of Hope, Duarte, California, USA; Center for Blood Cell Therapies at Peter McCallum Center, Melbourne, Australia; Departments of Molecular and Cellular Biology and Virology, City of Hope, Duarte, California, USA
| | - Elizabeth W Epps
- Shared Resources-Cellular Process, City of Hope, Duarte, California, USA; Center for Blood Cell Therapies at Peter McCallum Center, Melbourne, Australia; Departments of Molecular and Cellular Biology and Virology, City of Hope, Duarte, California, USA
| | - Nancy Gonzalez
- Shared Resources-Cellular Process, City of Hope, Duarte, California, USA; Center for Blood Cell Therapies at Peter McCallum Center, Melbourne, Australia; Departments of Molecular and Cellular Biology and Virology, City of Hope, Duarte, California, USA
| | - Chy-Anh Tran
- Shared Resources-Cellular Process, City of Hope, Duarte, California, USA; Center for Blood Cell Therapies at Peter McCallum Center, Melbourne, Australia; Departments of Molecular and Cellular Biology and Virology, City of Hope, Duarte, California, USA
| | - Xiwei Wu
- Shared Resources-Cellular Process, City of Hope, Duarte, California, USA; Center for Blood Cell Therapies at Peter McCallum Center, Melbourne, Australia; Departments of Molecular and Cellular Biology and Virology, City of Hope, Duarte, California, USA
| | - Jin-Hui Wang
- Shared Resources-Cellular Process, City of Hope, Duarte, California, USA; Center for Blood Cell Therapies at Peter McCallum Center, Melbourne, Australia; Departments of Molecular and Cellular Biology and Virology, City of Hope, Duarte, California, USA
| | - David L DiGiusto
- Shared Resources-Cellular Process, City of Hope, Duarte, California, USA; Center for Blood Cell Therapies at Peter McCallum Center, Melbourne, Australia; Departments of Molecular and Cellular Biology and Virology, City of Hope, Duarte, California, USA
| |
Collapse
|
36
|
Salvage therapy with everolimus reduces the severity of treatment-refractory chronic GVHD without impairing disease control: a dual center retrospective analysis. Bone Marrow Transplant 2014; 49:1412-8. [PMID: 25089598 DOI: 10.1038/bmt.2014.170] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 06/13/2014] [Accepted: 06/17/2014] [Indexed: 11/08/2022]
Abstract
Chronic GVHD (cGVHD) remains the most important cause of late non-relapse mortality post allogeneic hematopoietic SCT (HSCT). Although first-line treatment of cGVHD with steroids is well established, evidence for second-line treatment remains limited. Here, we report a dual center retrospective analysis of the off-label salvage treatment of steroid-refractory cGVHD with everolimus. Out of 80 patients with a median age of 50 (17-70) years, 14 (17%) suffered from mild, 39 (49%) from moderate and 27 (34%) from severe cGVHD. At the final analysis, median follow-up after introduction of everolimus was 724 (14-2205) days. Thirty-four patients (43%) required the addition of further immunosuppression during everolimus-based therapy. Global NIH Severity Score improved in 34 patients (43%), remained stable in 37 patients (46%) and worsened in 9 patients (11%). The total sum of Global NIH Severity Scores in all patients assessable was significantly reduced after treatment with everolimus (P<0.0001). Most frequent grade 3/4 toxicities included infections (n=30) and thrombocytopenia (n=15). There was a single case of relapse. Everolimus-based salvage treatment of refractory cGVHD results in significant improvement of the NIH Severity Score without impairing control of the malignant disease. Finally, these preliminary results demand further verification in prospective trials.
Collapse
|
37
|
Abstract
The last 6 decades have seen major advances in the understanding of immunologic diseases, driven by preclinical animal models. Indeed, bone marrow transplantation (BMT) has its genesis in rodent models dating back to the 1950s. Allogeneic BMT and its major complication, graft-versus-host disease (GVHD), represent a paradigm for the translation of preclinical concepts into clinical practice. The appreciation that GVHD can be thought of as a stepwise escalation in immune activation characterized by eventual massive target tissue apoptosis has allowed the design of rational approaches to better manage patients. Here, we describe the pathophysiology of GVHD as defined in preclinical models, focusing on the successes and failures of this research to instruct and translate clinical practice. We also provide a commentary on the limitations of these models so that they may be better appreciated and addressed in future studies. Notable preclinical successes include the definition of modern immune suppression, reductions in conditioning intensity, posttransplant cyclophosphamide, and the promotion of regulatory T-cell reconstitution. New strategies including naïve T-cell depletion, focused cytokine and chemokine inhibition, and the blockade of costimulation now also appear highly promising and very likely to translate into patients in the near future.
Collapse
|
38
|
Ram R, Storb R. Pharmacologic prophylaxis regimens for acute graft-versus-host disease: past, present and future. Leuk Lymphoma 2013; 54:1591-601. [PMID: 23278640 DOI: 10.3109/10428194.2012.762978] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Abstract Acute graft-versus-host disease (GVHD) has compromised and continues to compromise the benefits associated with allogeneic hematopoietic cell transplant to cure malignant and non-malignant diseases. Pharmacologic interventions to prevent GVHD have emerged as a major objective of research in the immunology and transplant fields. A better understanding of the pathobiology behind the GVHD process has led the way to novel approaches and medications. Here we review the present arsenal of medications used to prevent GVHD, focusing on past experience and the current evidence, and discuss future potential targets.
Collapse
Affiliation(s)
- Ron Ram
- Bone Marrow Transplantation Unit, Rabin Medical Center, Beilinson Hospital and the Sackler School of Medicine, Tel Aviv University, Israel.
| | | |
Collapse
|