51
|
A multicenter phase 1 study of nivolumab for relapsed hematologic malignancies after allogeneic transplantation. Blood 2021; 135:2182-2191. [PMID: 32478814 DOI: 10.1182/blood.2019004710] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 03/30/2020] [Indexed: 12/12/2022] Open
Abstract
Programmed cell death-1 (PD-1)/programmed death ligand-1 blockade may potentially augment graft-vs-tumor effects following allogeneic hematopoietic cell transplantation (alloHCT), but retrospective studies of anti-PD-1 therapy reported substantial toxicity from graft-versus-host-disease (GVHD). Here, we report the results of a prospective clinical trial of PD-1 blockade for relapsed hematologic malignancies (HMs) after alloHCT (NCT01822509). The primary objective in this phase 1 multicenter, investigator-initiated study was to determine maximum tolerated dose and safety. Secondary objectives were to assess efficacy and immunologic activity. Patients with relapsed HMs following alloHCT were eligible. Nivolumab was administered every 2 weeks until progression or unacceptable toxicity, starting with a 1-mg/kg cohort, with planned deescalation based on toxicity to a 0.5-mg/kg cohort. Twenty-eight patients were treated (n = 19 myeloid, n = 9 lymphoid). Median age was 57 years (range 27-76), and median time from alloHCT to enrollment was 21 months (range 5.6-108.5). Two of 6 patients treated at 1 mg/kg experienced dose-limiting toxicity (DLT) from immune-related adverse events (irAEs). Twenty-two patients were treated at 0.5 mg/kg, and 4 DLTs occurred, including 2 irAEs and 2 with fatal GVHD. The overall response rate in efficacy-evaluable patients was 32% (8/25). With a median follow-up of 11 months, the 1-year progression-free survival and overall survival were 23% and 56%, respectively. In this first prospective clinical trial of an anti-PD-1 antibody for post-alloHCT relapse, GVHD and irAEs occurred, requiring dose deescalation, with only modest antitumor activity. Further studies of anti-PD-1 therapy post-alloHCT may require specific toxicity mitigation strategies. This trial was registered at www.clinicaltrials.gov as #NCT01822509.
Collapse
|
52
|
Baliu-Piqué M, van Hoeven V, Drylewicz J, van der Wagen LE, Janssen A, Otto SA, van Zelm MC, de Boer RJ, Kuball J, Borghans JA, Tesselaar K. Cell-density independent increased lymphocyte production and loss rates post-autologous HSCT. eLife 2021; 10:59775. [PMID: 33538246 PMCID: PMC7886352 DOI: 10.7554/elife.59775] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 02/03/2021] [Indexed: 12/18/2022] Open
Abstract
Lymphocyte numbers need to be quite tightly regulated. It is generally assumed that lymphocyte production and lifespan increase homeostatically when lymphocyte numbers are low and, vice versa, return to normal once cell numbers have normalized. This widely accepted concept is largely based on experiments in mice, but is hardly investigated in vivo in humans. Here we quantified lymphocyte production and loss rates in vivo in patients 0.5–1 year after their autologous hematopoietic stem cell transplantation (autoHSCT). We indeed found that the production rates of most T- and B-cell subsets in autoHSCT-patients were two to eight times higher than in healthy controls, but went hand in hand with a threefold to ninefold increase in cell loss rates. Both rates also did not normalize when cell numbers did. This shows that increased lymphocyte production and loss rates occur even long after autoHSCT and can persist in the face of apparently normal cell numbers.
Collapse
Affiliation(s)
- Mariona Baliu-Piqué
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Vera van Hoeven
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Julia Drylewicz
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Anke Janssen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Sigrid A Otto
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Menno C van Zelm
- Department of Immunology and Pathology, Monash University and Alfred Hospital, Melbourne, Australia
| | - Rob J de Boer
- Theoretical Biology, Utrecht University, Utrecht, Netherlands
| | - Jürgen Kuball
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands.,Department of Hematology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jose Am Borghans
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Kiki Tesselaar
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
53
|
Rozmus J. Monogenic Immune Diseases Provide Insights Into the Mechanisms and Treatment of Chronic Graft-Versus-Host Disease. Front Immunol 2021; 11:574569. [PMID: 33613511 PMCID: PMC7889949 DOI: 10.3389/fimmu.2020.574569] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 12/07/2020] [Indexed: 12/22/2022] Open
Abstract
Chronic graft-versus-host disease (GvHD) has become a leading cause of morbidity and mortality following allogeneic hematopoietic stem cell transplantation (HSCT) and can burden patients with devastating and lifelong health effects. Our understanding of the pathogenic mechanisms underlying chronic GvHD remains incomplete and this lack of understanding is reflected by lack of clear therapeutic approaches to steroid refractory disease. Observations predominantly from mouse models and human correlative studies currently support a three phase model for the initiation and development of chronic GvHD: 1) early inflammation and tissue damage triggers the innate immune system. This leads to inflammatory cytokine/chemokine patterns that recruit effector immune cell populations; 2) chronic inflammation causes the loss of central and peripheral tolerance mechanisms leading to emergence of pathogenic B and T cell populations that promote autoimmune and alloimmune reactions; 3) the dysregulated immunity causes altered macrophage polarization, aberrant tissue repair leading to scarring and end organ fibrosis. This model has led to the evaluation of many new therapies aimed at limiting inflammation, targeting dysregulated signaling pathways and restoring tolerance mechanisms. However, chronic GvHD is a multisystem disease with complex clinical phenotypes and it remains unclear as to which cluster of patients will respond best to specific therapeutic strategies. However, it is possible to gain novel insights from immune-related monogenic diseases. These diseases either share common clinical manifestations, replicate steps from the three phase chronic GvHD model or serve as surrogates for perfectly targeted drugs being investigated in chronic GvHD therapy. In this review, we will summarize the evidence from these monogenic immune related diseases that provide insight into pathogenic pathways in chronic GvHD, rationales for current therapies and novel directions for future drug discovery.
Collapse
Affiliation(s)
- Jacob Rozmus
- Division of Pediatric Hematology, Oncology & BMT, Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada.,Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, Vancouver, BC, Canada
| |
Collapse
|
54
|
Rambaldi B, Kim HT, Reynolds C, Chamling Rai S, Arihara Y, Kubo T, Buon L, Gooptu M, Koreth J, Cutler C, Nikiforow S, Ho VT, Alyea EP, Antin JH, Wu CJ, Soiffer RJ, Ritz J, Romee R. Impaired T- and NK-cell reconstitution after haploidentical HCT with posttransplant cyclophosphamide. Blood Adv 2021; 5:352-364. [PMID: 33496734 PMCID: PMC7839379 DOI: 10.1182/bloodadvances.2020003005] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/25/2020] [Indexed: 12/18/2022] Open
Abstract
Administration of posttransplant cyclophosphamide (PTCy) has significantly expanded the number of patients undergoing HLA-haploidentical hematopoietic cell transplantation (haplo-HCT). To examine immune reconstitution in these patients, we monitored T- and natural killer (NK)-cell recovery in 60 patients receiving bone marrow or peripheral blood stem cell (PBSC) grafts after haplo-HCT with PTCy and 35 patients receiving HLA-matched donor PBSC grafts with standard graft-versus-host disease (GVHD) prophylaxis. Compared with HLA-matched recipients, early T-cell recovery was delayed in haplo-HCT patients and skewed toward effector memory T cells with markedly reduced naive T cells. We found higher regulatory T (Treg)-cell/conventional T (Tcon)-cell ratios early after HCT and increased PD-1 expression on memory T cells. Within the haplo-HCT, patients who did not develop chronic GVHD (cGVHD) had higher PD-1 expression on central and effector memory CD4+ Treg cells at 1 month after transplant. These findings suggest an immunologic milieu that promotes immune tolerance in haplo-HCT patients. NK cells were decreased early after haplo-HCT with preferential expansion of immature CD56brightCD16- NK cells compared with matched donor transplants. One month after transplant, mass cytometry revealed enrichment of immature NK-cell metaclusters with high NKG2A, low CD57, and low killer-cell immunoglobulin-like receptor expression after haplo-HCT, which partially recovered 3 months post-HCT. At 2 months, immature NK cells from both groups were functionally impaired, but interleukin-15 priming corrected these defects in vitro. Increased immature/mature NK-cell ratios were associated with cytomegalovirus reactivation and increased incidence of cGVHD after haplo-HCT. These homeostatic imbalances in T- and NK-cell reconstitution after haplo-HCT reveal opportunities for early immune-based interventions to optimize clinical outcomes.
Collapse
Affiliation(s)
- Benedetta Rambaldi
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
- Clinical and Experimental Sciences Department, Bone Marrow Transplant Unit, ASST Spedali Civili, University of Pavia, Brescia, Italy
| | - Haesook T Kim
- Department of Data Sciences, Dana-Farber Cancer Institute, Harvard T.H. Chan School of Public Health, Boston, MA; and
| | - Carol Reynolds
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Sharmila Chamling Rai
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Yohei Arihara
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Tomohiro Kubo
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Leutz Buon
- Department of BioInformatics and Data Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Mahasweta Gooptu
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - John Koreth
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Corey Cutler
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Sarah Nikiforow
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Vincent T Ho
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Edwin P Alyea
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Joseph H Antin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Robert J Soiffer
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Jerome Ritz
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Rizwan Romee
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| |
Collapse
|
55
|
Matos TR, Hirakawa M, Alho AC, Neleman L, Graca L, Ritz J. Maturation and Phenotypic Heterogeneity of Human CD4+ Regulatory T Cells From Birth to Adulthood and After Allogeneic Stem Cell Transplantation. Front Immunol 2021; 11:570550. [PMID: 33537026 PMCID: PMC7848157 DOI: 10.3389/fimmu.2020.570550] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 11/27/2020] [Indexed: 01/20/2023] Open
Abstract
CD4+ Regulatory T cells (Treg) play a critical role in maintaining immune homeostasis. Various Treg subsets have been identified, however the heterogeneity of Treg subpopulations during development remains uncharacterized. Using mass cytometry we obtained single cell data on expression of 35 functional markers to examine the heterogeneity of Treg cells at birth and in adults. Unsupervised clustering algorithms FlowSOM and ACCENSE were used to quantify Treg heterogeneity. As expected, Treg in umbilical cord blood were predominately naïve while Treg in adult blood were predominately central memory and effector memory cells. Although umbilical cord blood Treg are mostly naïve cells, we observed multiple phenotypic Treg subsets in cord blood. Nevertheless, peripheral blood in adults contained higher percentages of Treg and the heterogeneity of Treg was significantly increased in adults. We also studied Treg heterogeneity throughout a 2-year period after allogeneic hematopoietic stem cell transplantation (alloHSCT) and in patients with chronic graft-versus-host disease (cGVHD). Treg heterogeneity recovered rapidly after alloHSCT and gradually increased in the first two years post-transplant. However, patients with cGVHD had significantly fewer distinct Treg subpopulations, proposing a correlation between a disrupted Treg heterogeneity and cGVHD. Our study is the first to compare human Treg heterogeneity at birth, in healthy adults and in patients after alloHSCT with and without cGVHD. This approach to characterize Treg heterogeneity based on expression of a large panel of functional markers may enable future studies to identify specific Treg defects that contribute to immune dysfunction.
Collapse
Affiliation(s)
- Tiago R. Matos
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Amsterdam University Medical Centers, Department of Dermatology, University of Amsterdam, Amsterdam, Netherlands
| | - Masahiro Hirakawa
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Ana C. Alho
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Lars Neleman
- Amsterdam University Medical Centers, Department of Dermatology, University of Amsterdam, Amsterdam, Netherlands
| | - Luis Graca
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Jerome Ritz
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| |
Collapse
|
56
|
Bogunia-Kubik K, Łacina P. Non-KIR NK cell receptors: Role in transplantation of allogeneic haematopoietic stem cells. Int J Immunogenet 2020; 48:157-171. [PMID: 33352617 DOI: 10.1111/iji.12523] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/29/2020] [Accepted: 12/03/2020] [Indexed: 12/12/2022]
Abstract
Natural killer (NK) cells are of major significance in patients after allogeneic haematopoietic stem cell transplantation (HSCT). They are the first subset of lymphocytes to appear in peripheral blood after transplantation and play an important role in the immune responses against cancer and viral infections. The function of NK cells is controlled by various surface receptors, of which type I integral proteins with immunoglobulin-like domains (killer-cell immunoglobulin-like receptors, KIRs) have been the most extensively studied. The present review focuses on less studied NK cell receptors, such as type II integral proteins with lectin-like domains (CD94/NKG2, NKG2D), natural cytotoxicity receptors (NCRs), immunoglobulin-like transcripts (ILTs) and their ligands. Their potential role in patients with haematological disorders subjected to HSC transplant procedure in the context of post-transplant complications such as viral reactivation and acute graft-versus-host disease (GvHD) will be presented and discussed.
Collapse
Affiliation(s)
- Katarzyna Bogunia-Kubik
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Piotr Łacina
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| |
Collapse
|
57
|
Naserian S, Leclerc M, Shamdani S, Uzan G. Current Preventions and Treatments of aGVHD: From Pharmacological Prophylaxis to Innovative Therapies. Front Immunol 2020; 11:607030. [PMID: 33391276 PMCID: PMC7773902 DOI: 10.3389/fimmu.2020.607030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/16/2020] [Indexed: 12/16/2022] Open
Abstract
Graft versus host disease (GVHD) is one of the main causes of mortality and the reason for up to 50% of morbidity after hematopoietic stem cell transplantations (HSCT) which is the treatment of choice for many blood malignancies. Thanks to years of research and exploration, we have acquired a profound understanding of the pathophysiology and immunopathology of these disorders. This led to the proposition and development of many therapeutic approaches during the last decades, some of them with very promising results. In this review, we have focused on the recent GVHD treatments from classical chemical and pharmacological prophylaxis to more innovative treatments including gene therapy and cell therapy, most commonly based on the application of a variety of immunomodulatory cells. Furthermore, we have discussed the advantages and potentials of cell-free therapy as a newly emerging approach to treat GVHD. Among them, we have particularly focused on the implication of the TNFα-TNFR2 axis as a new immune checkpoint signaling pathway controlling different aspects of many immunoregulatory cells.
Collapse
Affiliation(s)
- Sina Naserian
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Villejuif, France
- Paris-Saclay University, Villejuif, France
- CellMedEx, Saint Maur Des Fossés, France
| | - Mathieu Leclerc
- Service d’Hématologie Clinique et de Thérapie Cellulaire, Hôpital Henri Mondor, Créteil, France
- INSERM U955, Institut Mondor de Recherche Biomédicale, Créteil, France
- Faculté de Médecine de Créteil, Université Paris-Est, Créteil, France
| | - Sara Shamdani
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Villejuif, France
- Paris-Saclay University, Villejuif, France
- CellMedEx, Saint Maur Des Fossés, France
| | - Georges Uzan
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Villejuif, France
- Paris-Saclay University, Villejuif, France
| |
Collapse
|
58
|
Wan L, Jin Z, Hu B, Lv K, Lei L, Liu Y, Song Y, Zhu Y, Gong H, Xu M, Du Y, Xu Y, Liu H, Wu D, Liu Y. IL-Y Aggravates Murine Chronic Graft- Versus-Host Disease by Enhancing T and B Cell Responses. Front Immunol 2020; 11:559740. [PMID: 33329519 PMCID: PMC7719702 DOI: 10.3389/fimmu.2020.559740] [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: 05/07/2020] [Accepted: 10/20/2020] [Indexed: 11/16/2022] Open
Abstract
IL-Y, a synthetic member of IL-12 cytokine family, was found to exert potent immunosuppressive effects by inhibiting the differentiation and activation of Th1 and Th17 cells. However, the role of IL-Y in the development of chronic graft-versus-host disease (cGVHD) remains unknown. Here, using murine models of scleroderma-like and lupus-like cGVHD, we examined the function of IL-Y in the pathogenesis of cGVHD by hydrodynamically injecting minicircle-IL-Y expressing plasmids (MC IL-Y). In contrast with the reported immune suppressive function of IL-Y, administration of MC IL-Y enhanced cGVHD severity reflected by deteriorated multi-organ pathologic damages. In lupus-like cGVHD model, urine protein and the serum anti-dsDNA antibody (IgG) were significantly upregulated by IL-Y treatment. Further study demonstrated that IL-Y impacts both donor T and B cell response. In T cells, IL-Y inhibited the generation of CD4+Foxp3+ regulator T (Treg) cells during the development of cGVHD. IL-Y may also increase the infiltration of pathogenic TNF-α producing CD4+ and CD8+ T cells through IL-27Rα in recipient spleens, as this effect was diminished in IL-27Rα deficient T cells. Moreover, IL-Y enhanced the differentiation of ICOS+ T follicular helper (Tfh) cells. In B cells, the percentage of germinal center (GC) B cells in recipient spleens was significantly upregulated by MC IL-Y plasmid administration. The levels of co-stimulatory molecules, MHC-II and CD86, on B cells were also enhanced by IL-Y expression. Taken together, our data indicated that IL-Y promoted the process of cGVHD by activating pathogenic T and B cells.
Collapse
Affiliation(s)
- Li Wan
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Ziqi Jin
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Bo Hu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Kangkang Lv
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Lei Lei
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yonghao Liu
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, Yoo Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yuan Song
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, Yoo Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ying Zhu
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, Yoo Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Huanle Gong
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Mimi Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yuanyuan Du
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yang Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Haiyan Liu
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, Yoo Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yuejun Liu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| |
Collapse
|
59
|
Allogeneic Hematopoietic Stem Cell Transplantation in Cutaneous T-Cell Lymphomas. Cancers (Basel) 2020; 12:cancers12102856. [PMID: 33023002 PMCID: PMC7601655 DOI: 10.3390/cancers12102856] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/30/2020] [Accepted: 10/01/2020] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Advanced-stage cutaneous T-cell lymphomas are aggressive diseases with frequent disease relapses and a reduced overall survival. Most treatment regimens fail to induce long-term remissions. Allogeneic hematopoietic stem cell transplantation has been associated with treatment-free long-term remissions and holds a potential for cure in this disease but is associated with frequent complications, mostly linked to the development of graft-versus-host disease and infections. Herein, we review the current evidence supporting the use of allogeneic stem cell transplantation in advanced-stage cutaneous T-cell lymphomas. Abstract Cutaneous T-cell lymphomas (CTCLs) are non-Hodgkin lymphomas that develop primarily in the skin. They account for almost 80% of primary cutaneous lymphomas. Epidermotropic CTCLs (mycosis fungoides (MF) and Sézary syndrome (SS)) are the most common form of CTCL. The course of the disease ranges from an indolent clinical behavior in early-stage disease to an aggressive evolution in the advanced stages. Advanced-stage disease is defined by the presence of tumors, erythroderma, or significant blood, nodal or visceral involvement. Advanced-stage disease is characterized by frequent disease relapses, refractory disease, a severely impaired quality of life and reduced overall survival. In the last twenty-five years, allogeneic hematopoietic stem cell transplantation (HSCT) has led to prolonged remissions in advanced CTCL, presumably linked to a graft-versus-lymphoma effect and is thus emerging as a potential cure of the disease. However, the high post-transplant relapse rate and severe morbidity and mortality associated with graft-versus-host disease and infections are important issues. Allogeneic HSCT is thus mostly considered in young patients with no comorbidities and an aggressive, advanced-stage CTCL. Allogeneic HSCT gives the best results in patients with a pre-transplant complete remission of the lymphoma. For this reason, one of the challenges is to define the best time to consider allogeneic HSCT in the disease course. Early identification of patients at high risk for progression is important to identify candidates who may benefit from allogeneic HSCT before their disease becomes treatment-refractory. This review describes the role of allogeneic HSCT in CTCL, summarizes the published data and future perspectives in this area.
Collapse
|
60
|
Dose-escalated interleukin-2 therapy for refractory chronic graft-versus-host disease in adults and children. Blood Adv 2020; 3:2550-2561. [PMID: 31471324 DOI: 10.1182/bloodadvances.2019000631] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 07/03/2019] [Indexed: 12/20/2022] Open
Abstract
Low-dose interleukin-2 (IL-2) therapy for chronic graft-versus-host disease (cGVHD) generates a rapid rise in plasma IL-2 levels and CD4+CD25+CD127-Foxp3+ regulatory T-cell (CD4Treg) proliferation, but both decrease over time despite continued daily administration. To test whether IL-2 dose escalation at the time of anticipated falls in plasma levels could circumvent tachyphylaxis and enhance CD4Treg expansion, we conducted a phase 1 trial in 10 adult and 11 pediatric patients with steroid-refractory cGVHD (www.clinicaltrials.gov: NCT02318082). Daily IL-2 was initiated in children and adults (0.33 × 106 and 0.67 × 106 IU/m2 per day, respectively). Dose escalations were scheduled at weeks 2 and 4 to a maximum dose of 1 × 106 IU/m2 per day in children and 2 × 106 IU/m2 per day in adults. Patients continued at their maximum tolerated dose (MTD) until week 8. Children tolerated IL-2 dose escalation with partial responses (PRs) in 9 of 11 patients (82%) at multiple cGVHD sites, including lung. Patient-reported outcome scores for skin and lung improved significantly in pediatric patients. In contrast, 5 of 10 adults required dose reduction, and only 2 of 7 evaluable patients (29%) had PRs at week 8. CD4Tregs and natural killer cells expanded in both cohorts without significant changes in conventional CD4+ T cells (Tcons) or CD8+ T cells. Children achieved a higher median CD4Treg/Tcon ratio at week 8 (0.4 vs 0.18, P = .02) despite lower IL-2 doses. We show for the first time that low-dose IL-2 is safe and effective in children with advanced cGVHD. In adults, escalation above the previously defined MTD did not improve CD4Treg expansion or clinical response.
Collapse
|
61
|
Fujiwara Y, Matsuoka KI, Iwamoto M, Sumii Y, Abe M, Mizuhara K, Urata T, Saeki K, Meguri Y, Asada N, Ennishi D, Nishimori H, Fujii K, Fujii N, Sugita J, Kobayashi H, Oto T, Maeda Y. Allogeneic hematopoietic stem cell transplantation in a prior lung transplant recipient. Int J Hematol 2020; 112:871-877. [PMID: 32803699 DOI: 10.1007/s12185-020-02967-x] [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: 04/23/2020] [Revised: 07/22/2020] [Accepted: 08/06/2020] [Indexed: 10/23/2022]
Abstract
Hematological diseases after solid organ transplant (SOT) are an emerging issue as the number of long-term SOT survivors increases. Expertise in managing patients requiring allogeneic hematopoietic stem cell transplantation (HSCT) after SOT from independent donors is needed; however, clinical reports of HSCT after SOT are limited, and the feasibility and risk are not well understood. In particular, HSCT in prior lung transplant recipients is thought to be complicated as the lung is immunologically distinct and is constantly exposed to the surrounding environment. Herein, we describe a case of successful HSCT in a patient with myelodysplastic syndromes who had previously received a lung transplant from a deceased donor for bronchiolitis obliterans syndrome. Reports about cases of HSCT after lung transplant are quite rare; thus, we discuss the mechanisms of immune tolerance through the clinical course of our case. This case suggests that HSCT after SOT can be considered a therapeutic option in cases where the transplanted organ is functionally retained and the hematological disease is in remission.
Collapse
Affiliation(s)
- Yuki Fujiwara
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikatacho, Kita-ku, Okayama-city, Okayama, 700-8558, Japan
| | - Ken-Ichi Matsuoka
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikatacho, Kita-ku, Okayama-city, Okayama, 700-8558, Japan.
| | - Miki Iwamoto
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikatacho, Kita-ku, Okayama-city, Okayama, 700-8558, Japan
| | - Yuichi Sumii
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikatacho, Kita-ku, Okayama-city, Okayama, 700-8558, Japan
| | - Masaya Abe
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikatacho, Kita-ku, Okayama-city, Okayama, 700-8558, Japan
| | - Kentaro Mizuhara
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikatacho, Kita-ku, Okayama-city, Okayama, 700-8558, Japan
| | - Tomohiro Urata
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikatacho, Kita-ku, Okayama-city, Okayama, 700-8558, Japan
| | - Kyosuke Saeki
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikatacho, Kita-ku, Okayama-city, Okayama, 700-8558, Japan
| | - Yusuke Meguri
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikatacho, Kita-ku, Okayama-city, Okayama, 700-8558, Japan
| | - Noboru Asada
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikatacho, Kita-ku, Okayama-city, Okayama, 700-8558, Japan
| | - Daisuke Ennishi
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikatacho, Kita-ku, Okayama-city, Okayama, 700-8558, Japan
| | - Hisakazu Nishimori
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikatacho, Kita-ku, Okayama-city, Okayama, 700-8558, Japan
| | - Keiko Fujii
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikatacho, Kita-ku, Okayama-city, Okayama, 700-8558, Japan
| | - Nobuharu Fujii
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikatacho, Kita-ku, Okayama-city, Okayama, 700-8558, Japan
| | - Junichi Sugita
- Department of Hematology, Hokkaido University Hospital, Hokkaido, Japan
| | - Hajime Kobayashi
- Department of Hematology, Obihiro Kosei Hospital, Hokkaido, Japan
| | - Takahiro Oto
- Department of Organ Transplant Center, Okayama University Hospital, Okayama, Japan
| | - Yoshinobu Maeda
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikatacho, Kita-ku, Okayama-city, Okayama, 700-8558, Japan
| |
Collapse
|
62
|
Functional analysis of clinical response to low-dose IL-2 in patients with refractory chronic graft-versus-host disease. Blood Adv 2020; 3:984-994. [PMID: 30936059 DOI: 10.1182/bloodadvances.2018027474] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 02/19/2019] [Indexed: 12/13/2022] Open
Abstract
Patients with chronic graft-versus-host disease (cGVHD) have a paucity of regulatory CD4 T cells (CD4Tregs) that mediate peripheral tolerance. In clinical trials, daily low-dose interleukin-2 (IL-2) has been administered safely for prolonged periods in patients with steroid-refractory cGVHD. Peripheral CD4Tregs expand dramatically in all patients during IL-2 therapy but clinical improvement was observed in ∼50% of patients. Here, we examined the impact of low-dose IL-2 therapy on functional T-cell markers and the T-cell repertoire within CD4Tregs, conventional CD4 T cells (CD4Tcons), and CD8+ T cells. IL-2 had profound effects on CD4Tregs homeostasis in both response groups including selective expansion of the naive subset, improved thymic output, and increased expression of Ki67, FOXP3, and B-cell lymphoma 2 within CD4Tregs. Similar changes were not seen in CD4Tcons or CD8 T cells. Functionally, low-dose IL-2 enhanced, in vitro, CD4Treg-suppressive activity in both response groups, and all patient CD4Tcons were similarly suppressed by healthy donor CD4Tregs. High-throughput sequencing of the T-cell receptor β (TCRβ) locus demonstrated that low-dose IL-2 therapy increased TCR repertoire diversity and decreased evenness within CD4Tregs without affecting CD4Tcons or CD8 T cells. Using clone-tracking analysis, we observed rapid turnover of highly prevalent clones in CD4Tregs as well as the conversion of CD4Tcons to CD4Tregs. After 12 weeks of daily IL-2, clinical responders had a greater influx of novel clones within the CD4Treg compartment compared with nonresponders. Further studies to define the function and specificity of these novel CD4Treg clones may help establish the mechanisms whereby low-dose IL-2 therapy promotes immune tolerance.
Collapse
|
63
|
Dekker L, de Koning C, Lindemans C, Nierkens S. Reconstitution of T Cell Subsets Following Allogeneic Hematopoietic Cell Transplantation. Cancers (Basel) 2020; 12:E1974. [PMID: 32698396 PMCID: PMC7409323 DOI: 10.3390/cancers12071974] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/10/2020] [Accepted: 07/16/2020] [Indexed: 02/07/2023] Open
Abstract
Allogeneic (allo) hematopoietic cell transplantation (HCT) is the only curative treatment option for patients suffering from chemotherapy-refractory or relapsed hematological malignancies. The occurrence of morbidity and mortality after allo-HCT is still high. This is partly correlated with the immunological recovery of the T cell subsets, of which the dynamics and relations to complications are still poorly understood. Detailed information on T cell subset recovery is crucial to provide tools for better prediction and modulation of adverse events. Here, we review the current knowledge regarding CD4+ and CD8+ T cells, γδ T cells, iNKT cells, Treg cells, MAIT cells and naive and memory T cell reconstitution, as well as their relations to outcome, considering different cell sources and immunosuppressive therapies. We conclude that the T cell subsets reconstitute in different ways and are associated with distinct adverse and beneficial events; however, adequate reconstitution of all the subsets is associated with better overall survival. Although the exact mechanisms involved in the reconstitution of each T cell subset and their associations with allo-HCT outcome need to be further elucidated, the data and suggestions presented here point towards the development of individualized approaches to improve their reconstitution. This includes the modulation of immunotherapeutic interventions based on more detailed immune monitoring, aiming to improve overall survival changes.
Collapse
Affiliation(s)
- Linde Dekker
- Princess Máxima Center for Pediatric Oncology, Utrecht University, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (L.D.); (C.L.)
| | - Coco de Koning
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands;
| | - Caroline Lindemans
- Princess Máxima Center for Pediatric Oncology, Utrecht University, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (L.D.); (C.L.)
| | - Stefan Nierkens
- Princess Máxima Center for Pediatric Oncology, Utrecht University, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (L.D.); (C.L.)
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands;
| |
Collapse
|
64
|
Gonzalez RM, Pidala J. Evolving Therapeutic Options for Chronic Graft‐versus‐Host Disease. Pharmacotherapy 2020; 40:756-772. [DOI: 10.1002/phar.2427] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 04/04/2020] [Accepted: 04/13/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Rebecca M. Gonzalez
- Department of Blood & Marrow Transplant and Cellular Immunotherapy (BMT CI) Moffitt Cancer Center Tampa Florida USA
- Department of Pharmacy Moffitt Cancer Center Tampa Florida USA
| | - Joseph Pidala
- Department of Blood & Marrow Transplant and Cellular Immunotherapy (BMT CI) Moffitt Cancer Center Tampa Florida USA
| |
Collapse
|
65
|
Talvard-Balland N, Sutra Del Galy A, Michonneau D, Le Buanec H, Chasset F, Robin M, Peffault de Latour R, Xhaard A, Sicre de Fontbrune F, Parquet N, Duchez S, Schiavon V, Rybojad M, Bergeron-Lafaurie A, Bagot M, Bensussan A, Caillat-Zucman S, Socié G, Bouaziz JD, de Masson A. Expansion of Circulating CD49b +LAG3 + Type 1 Regulatory T Cells in Human Chronic Graft-Versus-Host Disease. J Invest Dermatol 2020; 141:193-197.e2. [PMID: 32428542 DOI: 10.1016/j.jid.2020.04.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/27/2020] [Accepted: 04/24/2020] [Indexed: 12/21/2022]
Affiliation(s)
- Nana Talvard-Balland
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France; Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France
| | - Aurélien Sutra Del Galy
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France; Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France; Service d'Hématologie-Greffes, AP-HP, Hôpital Saint-Louis, Paris, France
| | - David Michonneau
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France; Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France; Service d'Hématologie-Greffes, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Helene Le Buanec
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France; Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France
| | - Francois Chasset
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France; Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France; Service de Dermatologie, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Marie Robin
- Service d'Hématologie-Greffes, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Régis Peffault de Latour
- Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France; Service d'Hématologie-Greffes, AP-HP, Hôpital Saint-Louis, Paris, France; INSERM UMRS 1160, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France
| | - Alienor Xhaard
- Service d'Hématologie-Greffes, AP-HP, Hôpital Saint-Louis, Paris, France
| | | | - Nathalie Parquet
- Service d'Aphérèse Thérapeutique, AP-HP, Hôpital Saint-Louis, France
| | - Sophie Duchez
- UMR CNRS 7212, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France
| | - Valérie Schiavon
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France
| | - Michel Rybojad
- Service de Dermatologie, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Anne Bergeron-Lafaurie
- Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France; Service de Pneumologie, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Martine Bagot
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France; Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France; Service de Dermatologie, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Armand Bensussan
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France; Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France
| | - Sophie Caillat-Zucman
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France; Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France; Laboratoire d'Immunologie, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Gérard Socié
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France; Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France; Service d'Hématologie-Greffes, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Jean-David Bouaziz
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France; Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France; Service de Dermatologie, AP-HP, Hôpital Saint-Louis, Paris, France.
| | - Adèle de Masson
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France; Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, Paris, France; Service de Dermatologie, AP-HP, Hôpital Saint-Louis, Paris, France
| |
Collapse
|
66
|
Woelfinger P, Epp K, Schaefer L, Kriege D, Theobald M, Bopp T, Wagner-Drouet EM. CD52-negative T cells predict acute graft-versus-host disease after an alemtuzumab-based conditioning regimen. Br J Haematol 2020; 191:253-262. [PMID: 32410220 DOI: 10.1111/bjh.16706] [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] [Received: 01/16/2020] [Accepted: 04/09/2020] [Indexed: 11/28/2022]
Abstract
Allogeneic haematopoietic stem cell transplantation (HSCT) after a reduced-intensity conditioning (RIC) regimen with fludarabine, melphalan and alemtuzmab is an effective therapy for haematological malignancies. Alemtuzumab, a monoclonal antibody against CD52, a glycosylphosphatidylinositol-anchor-bound surface protein on lymphocytes, depletes T cells to prevent graft-versus-host disease (GVHD). Despite this, acute and chronic GVHD (a/cGVHD) remain life-threatening complications after HSCT. The aim of the present study was to identify parameters to predict GVHD. In 69 patients after HSCT, T-cell subsets were functionally analysed. Reconstitution of CD52neg T cells and CD52neg regulatory T cells (Tregs) correlated with onset, severity and clinical course of aGVHD. Patients with aGVHD showed significantly lower levels of CD52pos T cells compared to patients with cGVHD or without GVHD (P < 0·001). Analysis of T-cell reconstitution revealed a percentage of <40% of CD52pos CD4pos T cells or CD52pos Tregs at day +50 as a risk factor for the development of aGVHD. In contrast, CD52neg Tregs showed significant decreased levels of glycoprotein A repetitions predominant (GARP; P < 0·001), glucocorticoid-induced TNFR-related protein (GITR; P < 0·001), chemokine receptor (CXCR3; P = 0·023), C-C chemokine receptor type 5 (CCR5; P = 0·004), but increased levels of immunoglobulin-like transcript 3 (ILT3; P = 0·001), as well as a reduced suppressive capacity. We conclude that reconstitution of CD52neg T cells and CD52neg Tregs is a risk factor for development of aGVHD.
Collapse
Affiliation(s)
- Pascal Woelfinger
- Department of Hematology, Oncology and Pneumology, University Cancer Center Mainz (UCT), University Medical Center Mainz, Mainz, Germany
| | - Katharina Epp
- Department of Hematology, Oncology and Pneumology, University Cancer Center Mainz (UCT), University Medical Center Mainz, Mainz, Germany
| | - Lukas Schaefer
- Department of Hematology, Oncology and Pneumology, University Cancer Center Mainz (UCT), University Medical Center Mainz, Mainz, Germany
| | - Diana Kriege
- Department of Hematology, Oncology and Pneumology, University Cancer Center Mainz (UCT), University Medical Center Mainz, Mainz, Germany
| | - Matthias Theobald
- Department of Hematology, Oncology and Pneumology, University Cancer Center Mainz (UCT), University Medical Center Mainz, Mainz, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Eva-Maria Wagner-Drouet
- Department of Hematology, Oncology and Pneumology, University Cancer Center Mainz (UCT), University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
67
|
Zhang X, Olsen N, Zheng SG. The progress and prospect of regulatory T cells in autoimmune diseases. J Autoimmun 2020; 111:102461. [PMID: 32305296 DOI: 10.1016/j.jaut.2020.102461] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 04/08/2020] [Indexed: 12/16/2022]
Abstract
Regulatory T cells (Treg) are an important immune cell population, playing a crucial role in regulating immune tolerance and preventing autoimmune diseases. These cells consist of various cell sub-populations and generally have an immunoregulatory or suppressive role against immune responses. They also have a different cell heterogeneity and each populations has own biological characteristics. Treg deficiency, reduction, instability, reduced vitality and dysfunction all account for multiple autoimmune diseases. In this review, we have systemically reviewed Treg classification, phenotypic features, regulation of Foxp3 expression, plasticity and stability of Treg as well as their relationship with several important autoimmune diseases. We particularly focus on why and how inflammatory and diet environments affect the functional capacity and underlying mechanisms of Treg cell populations. We also summarize new advances in technologies which help to analyze and dissect these cells in molecular levels in-depth. We also clarify the possible clinical relevance on application of these cells in patients with autoimmune diseases. The advantages and weaknesses have been carefully discussed as well. We also propose the possible approaches to overcome these weaknesses of Treg cells in complicate environments. Thus, we have displayed the updated knowledge of Treg cells, which provides an overall insight into the role and mechanisms of Treg cells in autoimmune diseases.
Collapse
Affiliation(s)
- Ximei Zhang
- Institute of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China; Division of Rheumatology and Immunology, Department of Internal Medicine, Ohio State University College of Medicine, Columbus, PA, 43201, USA
| | - Nancy Olsen
- Division of Rheumatology, Department of Medicine at Penn State College of Medicine and Milton S. Hershey Medical Center, Hershey, 17033, USA
| | - Song Guo Zheng
- Division of Rheumatology and Immunology, Department of Internal Medicine, Ohio State University College of Medicine, Columbus, PA, 43201, USA.
| |
Collapse
|
68
|
Schultz KR, Kariminia A, Ng B, Abdossamadi S, Lauener M, Nemecek ER, Wahlstrom JT, Kitko CL, Lewis VA, Schechter T, Jacobsohn DA, Harris AC, Pulsipher MA, Bittencourt H, Choi SW, Caywood EH, Kasow KA, Bhatia M, Oshrine BR, Flower A, Chaudhury S, Coulter D, Chewning JH, Joyce M, Savasan S, Pawlowska AB, Megason GC, Mitchell D, Cheerva AC, Lawitschka A, Azadpour S, Ostroumov E, Subrt P, Halevy A, Mostafavi S, Cuvelier GDE. Immune profile differences between chronic GVHD and late acute GVHD: results of the ABLE/PBMTC 1202 studies. Blood 2020; 135:1287-1298. [PMID: 32047896 PMCID: PMC7146024 DOI: 10.1182/blood.2019003186] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 01/20/2020] [Indexed: 12/14/2022] Open
Abstract
Human graft-versus-host disease (GVHD) biology beyond 3 months after hematopoietic stem cell transplantation (HSCT) is complex. The Applied Biomarker in Late Effects of Childhood Cancer study (ABLE/PBMTC1202, NCT02067832) evaluated the immune profiles in chronic GVHD (cGVHD) and late acute GVHD (L-aGVHD). Peripheral blood immune cell and plasma markers were analyzed at day 100 post-HSCT and correlated with GVHD diagnosed according to the National Institutes of Health consensus criteria (NIH-CC) for cGVHD. Of 302 children enrolled, 241 were evaluable as L-aGVHD, cGVHD, active L-aGVHD or cGVHD, and no cGVHD/L-aGVHD. Significant marker differences, adjusted for major clinical factors, were defined as meeting all 3 criteria: receiver-operating characteristic area under the curve ≥0.60, P ≤ .05, and effect ratio ≥1.3 or ≤0.75. Patients with only distinctive features but determined as cGVHD by the adjudication committee (non-NIH-CC) had immune profiles similar to NIH-CC. Both cGVHD and L-aGVHD had decreased transitional B cells and increased cytolytic natural killer (NK) cells. cGVHD had additional abnormalities, with increased activated T cells, naive helper T (Th) and cytotoxic T cells, loss of CD56bright regulatory NK cells, and increased ST2 and soluble CD13. Active L-aGVHD before day 114 had additional abnormalities in naive Th, naive regulatory T (Treg) cell populations, and cytokines, and active cGVHD had an increase in PD-1- and a decrease in PD-1+ memory Treg cells. Unsupervised analysis appeared to show a progression of immune abnormalities from no cGVHD/L-aGVHD to L-aGVHD, with the most complex pattern in cGVHD. Comprehensive immune profiling will allow us to better understand how to minimize L-aGVHD and cGVHD. Further confirmation in adult and pediatric cohorts is needed.
Collapse
Affiliation(s)
- Kirk R Schultz
- Michael Cuccione Childhood Cancer research program, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Amina Kariminia
- Michael Cuccione Childhood Cancer research program, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Bernard Ng
- Department of Statistics, Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Sayeh Abdossamadi
- Michael Cuccione Childhood Cancer research program, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Madeline Lauener
- Michael Cuccione Childhood Cancer research program, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Eneida R Nemecek
- Pediatric Blood and Marrow Transplantation, Doernbecher Children's Hospital, Oregon Health and Science University, Portland, OR
| | - Justin T Wahlstrom
- Blood and Marrow Transplantation Program, Benioff Children's Hospital, University of California San Francisco, San Francisco, CA
| | - Carrie L Kitko
- Pediatric Stem Cell Transplantation Program, Vanderbilt University Medical Center, Nashville, TN
| | - Victor A Lewis
- Pediatric Oncology, Alberta Children's Hospital, University of Calgary, Calgary, AB, Canada
| | - Tal Schechter
- Hematology-Oncology, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - David A Jacobsohn
- Blood and Marrow Transplantation, Children's National Health System, Washington, DC
| | - Andrew C Harris
- Pediatric Hematology Oncology, Primary Children's Hospital, University of Utah, Salt Lake City, UT
| | - Michael A Pulsipher
- Blood and Marrow Transplantation, Children's Hospital Los Angeles, Los Angeles, CA
| | - Henrique Bittencourt
- Hematology Oncology, Sainte-Justine University Hospital Center, Montreal, QC, Canada
| | - Sung Won Choi
- Michigan Medicine Pediatric Bone Marrow Transplant, C.S. Mott Children's Hospital, Michigan Medicine, Ann Arbor, MI
| | - Emi H Caywood
- Pediatric Hematology Oncology, Nemours Alfred I. duPont Hospital for Children, Wilmington, DE
| | - Kimberly A Kasow
- Pediatric Bone Marrow Transplant, University of North Carolina, Chapel Hill, NC
| | - Monica Bhatia
- Pediatric Stem Cell Transplant Program, Morgan Stanley Children's Hospital, Columbia University, New York, NY
| | - Benjamin R Oshrine
- Oncology and Hematology, Johns Hopkins All Children's Hospital, St. Petersburg, FL
| | - Allyson Flower
- Division of Pediatric Hematology, Oncology, Stem Cell Transplant, New York Medical College, Valhalla, NY
| | - Sonali Chaudhury
- Hematology, Oncology, Neuro-Oncology & Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital, Northwestern University, Chicago, IL
| | - Donald Coulter
- Division of Pediatric Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE
| | - Joseph H Chewning
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL
| | - Michael Joyce
- Division of Pediatric Hematology/Oncology Clinic, Nemours Children's Specialty Care, Jacksonville, FL
| | - Sureyya Savasan
- Pediatric Hematology & Oncology, Children's Hospital of Michigan, Detroit, MI
| | | | - Gail C Megason
- Children's Hematology/Oncology, University of Mississippi Medical Center, Jackson, MS
| | - David Mitchell
- Division of Pediatric Hematology/Oncology, Montreal Children's Hospital, Montreal, QC
| | - Alexandra C Cheerva
- Pediatric Hematology, Oncology and Stem Cell Transplantation, Norton Children's Hospital, University of Louisville, Louisville, KY
| | - Anita Lawitschka
- Stem Cell Transplant Outpatient & Aftercare Clinic, St. Anna Children's Hospital, Medical University Vienna, Vienna, Austria
| | | | - Elena Ostroumov
- Michael Cuccione Childhood Cancer research program, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Peter Subrt
- Michael Cuccione Childhood Cancer research program, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Anat Halevy
- Michael Cuccione Childhood Cancer research program, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Sara Mostafavi
- Department of Statistics, Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
69
|
Strobl J, Pandey RV, Krausgruber T, Kleissl L, Reininger B, Herac M, Bayer N, Krall C, Wohlfarth P, Mitterbauer M, Kalhs P, Rabitsch W, Bock C, Hopfinger G, Stary G. Anti-Apoptotic Molecule BCL2 Is a Therapeutic Target in Steroid-Refractory Graft-Versus-Host Disease. J Invest Dermatol 2020; 140:2188-2198. [PMID: 32247860 DOI: 10.1016/j.jid.2020.02.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/11/2020] [Accepted: 02/23/2020] [Indexed: 12/12/2022]
Abstract
Graft-versus-host disease (GVHD) is the leading cause of mortality after hematopoietic stem cell transplantation and primarily affects barrier organs such as the skin. One-third of cases are refractory to steroid treatment resulting in poor outcomes and the need for novel therapies. Longitudinal analysis of T-cell transcriptomes in patients before the appearance of GVHD symptoms revealed the upregulation of anti-apoptotic regulator B-cell lymphoma 2 (BCL2) at GVHD initiation. To determine the potential of BCL2 inhibition in active GVHD, we analyzed tissues of 88 patients with acute or chronic GVHD. BCL2 RNA was elevated in multiple organs affected by GVHD and expression correlated with transplant-related mortality and steroid-refractory GVHD. BCL2-expressing lymphocytes were present in skin lesions and peripheral blood of patients with acute and chronic GVHD. Inhibition of BCL2 increased the CD4 to CD8 ratio in allogeneic T cells in vitro and induced apoptosis of T cells from patients with steroid-pretreated chronic GVHD ex vivo. In addition, the higher ratio of regulatory to nonregulatory T cells upon blockage of BCL2 could add to the anti-inflammatory effect of BCL2 blockage. Collectively, our results highlight BCL2 as an important factor for GVHD development and introduce BCL2 inhibition as previously unreported and urgently needed targeted therapy in the treatment of steroid-refractory GVHD.
Collapse
Affiliation(s)
- Johanna Strobl
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Ram Vinay Pandey
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Thomas Krausgruber
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Lisa Kleissl
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Bärbel Reininger
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Merima Herac
- Clinical Institute for Pathology, Medical University of Vienna, Vienna, Austria
| | - Nadine Bayer
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Christoph Krall
- Center for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna, Austria
| | - Philipp Wohlfarth
- Department of Internal Medicine I, Bone Marrow Transplantation Unit, Medical University of Vienna, Vienna, Austria
| | - Margit Mitterbauer
- Department of Internal Medicine I, Bone Marrow Transplantation Unit, Medical University of Vienna, Vienna, Austria
| | - Peter Kalhs
- Department of Internal Medicine I, Bone Marrow Transplantation Unit, Medical University of Vienna, Vienna, Austria
| | - Werner Rabitsch
- Department of Internal Medicine I, Bone Marrow Transplantation Unit, Medical University of Vienna, Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Georg Hopfinger
- Department of Internal Medicine I, Bone Marrow Transplantation Unit, Medical University of Vienna, Vienna, Austria
| | - Georg Stary
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.
| |
Collapse
|
70
|
Goshima Y, Nakaoka S, Ohashi K, Sakamaki H, Shibuya K, Shibuya A. A mathematical model for dynamics of soluble form of DNAM-1 as a biomarker for graft-versus-host disease. PLoS One 2020; 15:e0228508. [PMID: 32040515 PMCID: PMC7010286 DOI: 10.1371/journal.pone.0228508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 01/16/2020] [Indexed: 12/29/2022] Open
Abstract
DNAM-1 (CD226) is an activating immunoreceptor expressed on T cells and NK cells and involved in the pathogenesis of acute graft-versus-host disease (aGVHD) after allogeneic hematopoietic stem cell transplantation (allo-HSCT). We previously reported that a soluble form of DNAM-1 (sDNAM-1) is generated by shedding from activated T cells. Moreover, higher serum levels of sDNAM-1 in patients before allo-HSCT is a predictive biomarker for the development of aGVHD based on the retrospective univariate and multivariate analyses in allo-HSCT patients. However, it remains unclear how the serum levels of sDNAM-1 are regulated after allo-HSCT and whether they are associated with the development of aGVHD. Here, we constructed a mathematical model to assess the dynamics of sDNAM-1 after allo-HSCT by assuming that there are three types of sDNAM-1 (the first and the second were from alloreactive and non-alloreactive donor lymphocytes, respectively, and the third from recipient lymphocytes). Our mathematical model fitted well to the data set of sDNAM-1 in patients (n = 67) who had undergone allo-HSCT and suggest that the high proportion of the first type of sDNAM-1 to the total of the first and second types is associated with high risk of the development of severe aGVHD. Thus, sDNAM-1 after allo-HSCT can be a biomarker for the development of aGVHD.
Collapse
Affiliation(s)
- Yuki Goshima
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- School of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Shinji Nakaoka
- Faculty of Advanced Life Science, Hokkaido University, Sapporo, Hokkaido, Japan
- PRESTO, Japan Science and Technology Agency, Saitama, Japan
| | - Kazuteru Ohashi
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Hisashi Sakamaki
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Kazuko Shibuya
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akira Shibuya
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
- * E-mail:
| |
Collapse
|
71
|
Whangbo JS, Antin JH, Koreth J. The role of regulatory T cells in graft-versus-host disease management. Expert Rev Hematol 2020; 13:141-154. [PMID: 31874061 DOI: 10.1080/17474086.2020.1709436] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: Despite improvements in human leukocyte antigen (HLA) matching algorithms and supportive care, graft-versus-host disease (GVHD) remains the leading cause of non-relapse morbidity and mortality following allogeneic hematopoietic stem cell transplantation (HSCT). Acute GVHD, typically occurring in the first 100 days post-HSCT, is mediated by mature effector T cells from the donor (graft) that become activated after encountering alloantigens in the recipient (host). Chronic GVHD, characterized by aberrant immune responses to both autoantigens and alloantigens, occurs later and arises from a failure to develop tolerance after HSCT. CD4+ CD25+ CD127- FOXP3+ regulatory T cells (Tregs) function to suppress auto- and alloreactive immune responses and are key mediators of immune tolerance.Areas covered: In this review, authors discuss the biologic and therapeutic roles of Tregs in acute and chronic GVHD, including in vivo and ex vivo strategies for Treg expansion and adoptive Treg cellular therapy.Expert opinion: Although they comprise only a small subset of circulating CD4 + T cells, Tregs play an important role in establishing and maintaining immune tolerance following allogeneic HSCT. The development of GVHD has been associated with reduced Treg frequency or numbers. Consequently, the immunosuppressive properties of Tregs are being harnessed in clinical trials for GVHD prevention and treatment.
Collapse
Affiliation(s)
- Jennifer S Whangbo
- Division of Hematology-Oncology, Boston Children's Hospital, Boston, MA and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Joseph H Antin
- Harvard Medical School, Boston, MA, USA.,Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA, USA
| | - John Koreth
- Harvard Medical School, Boston, MA, USA.,Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
72
|
Yoshida H, Koike M, Tada Y, Nakata K, Hino A, Fuji S, Masaie H, Oka C, Higeno A, Idota A, Yamasaki T, Ishikawa J. Different Immune Reconstitution between Cord Blood and Unrelated Bone Marrow Transplantation with Relation to Chronic Graft-versus-Host Disease. Int J Hematol Oncol Stem Cell Res 2020; 14:1-10. [PMID: 32337009 PMCID: PMC7167606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Background: Advances of allogeneic hematopoietic cell transplantation (allo-HCT) have brought long-term survival to the patients with hematologic malignancies. Chronic graft-versus-host disease (GVHD) is one of major problems for the long- term survivors after allo-HCT. Dysregulation of immune reconstitution has been reported to be involved in the pathogenesis of chronic GVHD. Differences of immune reconstitution between cord blood transplantation (CBT) and unrelated bone marrow transplantation (UBMT) remain unclear in long-term survivors. We investigated immune reconstitution in patients surviving for more than 2 years after CBT (n=21) or UBMT (n=20) without relapse of underlying disease. Materials and Methods: Using flow cytometric analysis of peripheral blood, we investigated immune reconstitution of T cells, B cells, and NK cells between CBT and UBMT patients. We collected clinical data regarding allo-HCT and examined the relation of immune reconstitution to the development of chronic GVHD. Results: Between CBT and UBMT patients, we found significant differences in absolute cell number of CD8+ as well as CD19+ cell and CD4/CD8 ratio even more than 2 years after allo-HCT. Among UBMT patients, absolute cell number of naive CD4+ cell was significantly lower in patients with chronic GVHD. In addition, we found significant differences in absolute cell number of CD19+ cell, especially naive B cell between patients with and without chronic GVHD in both CBT and UBMT patients. Conclusion: These results suggest that differences of immune recovery between CBT and UBMT patients may exist even in patients surviving for more than 2 years and might be related to the development of chronic GVHD.
Collapse
Affiliation(s)
- Hitoshi Yoshida
- Department of Hematology, Osaka International Cancer Institute, Osaka, Japan
| | - Midori Koike
- Department of Hematology, Osaka International Cancer Institute, Osaka, Japan
| | - Yuma Tada
- Department of Hematology, Osaka International Cancer Institute, Osaka, Japan
| | - Keiichi Nakata
- Department of Hematology, Osaka International Cancer Institute, Osaka, Japan
| | - Akihisa Hino
- Department of Hematology, Osaka International Cancer Institute, Osaka, Japan
| | - Shigeo Fuji
- Department of Hematology, Osaka International Cancer Institute, Osaka, Japan
| | - Hiroaki Masaie
- Department of Hematology, Osaka International Cancer Institute, Osaka, Japan
| | - Chihiro Oka
- Department of Laboratory, Osaka International Cancer Institute, Osaka, Japan
| | - Akemi Higeno
- Department of Laboratory, Osaka International Cancer Institute, Osaka, Japan
| | - Atsushi Idota
- Department of Laboratory, Osaka International Cancer Institute, Osaka, Japan
| | - Tomoyuki Yamasaki
- Department of Laboratory, Osaka International Cancer Institute, Osaka, Japan
| | - Jun Ishikawa
- Department of Hematology, Osaka International Cancer Institute, Osaka, Japan
| |
Collapse
|
73
|
Iamsawat S, Tian L, Daenthanasanmak A, Wu Y, Nguyen HD, Bastian D, Yu XZ. Vitamin C stabilizes CD8+ iTregs and enhances their therapeutic potential in controlling murine GVHD and leukemia relapse. Blood Adv 2019; 3:4187-4201. [PMID: 31856270 PMCID: PMC6929397 DOI: 10.1182/bloodadvances.2019000531] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 11/14/2019] [Indexed: 02/06/2023] Open
Abstract
Adoptive transfer of induced regulatory T cells (iTregs) can ameliorate graft-versus-host disease (GVHD) after allogeneic hematopoietic cell transplantation (allo-HCT). CD4+ iTregs can effectively prevent GVHD but impair the graft-versus-leukemia (GVL) effect, whereas CD8+ iTregs preserve the GVL effect but have limited efficacy in GVHD control because of their instability under inflammatory conditions. Thus, we aimed to stabilize CD8+ iTregs via treatment with vitamin C (Vit C) to improve their efficacy in controlling GVHD. We found that addition of Vit C significantly improved the stability of forkhead box P3 (Foxp3) expression in CD8+ iTregs. Moreover, Vit C-treated CD8+ iTregs exhibited high efficacy in attenuating acute and chronic GVHD. The mechanistic study revealed that addition of Vit C to CD8+ iTreg culture markedly increased DNA demethylation in the conserved noncoding sequence 2 region and, hence, maintained higher Foxp3 expression levels compared with untreated controls. In acute GVHD, Vit C-treated CD8+ iTregs were able to inhibit pathogenic T-cell expansion and differentiation while reducing thymus damage and B-cell activation in cGVHD. Importantly, in contrast to CD4+ iTregs, Vit C-treated CD8+ iTregs retained the ability to control tumor relapse. These results provide a strong rationale to use Vit C in the clinic to stabilize CD8+ iTregs for the control of GVHD and preservation of GVL after allo-HCT.
Collapse
Affiliation(s)
| | - Linlu Tian
- Department of Microbiology and Immunology and
| | | | - Yongxia Wu
- Department of Microbiology and Immunology and
| | | | | | - Xue-Zhong Yu
- Department of Microbiology and Immunology and
- Department of Medicine, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
74
|
Zhang P, Yang S, Zou Y, Yan X, Wu H, Zhou M, Sun YC, Zhang Y, Zhu H, Xu K, Wang Y, Sheng LX, Mu Q, Sun L, Ouyang G. NK cell predicts the severity of acute graft-versus-host disease in patients after allogeneic stem cell transplantation using antithymocyte globulin (ATG) in pretreatment scheme. BMC Immunol 2019; 20:46. [PMID: 31818250 PMCID: PMC6902350 DOI: 10.1186/s12865-019-0326-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/18/2019] [Indexed: 12/29/2022] Open
Abstract
Background Graft-versus-host disease (GVHD) is one of the most complex complications after allogeneic stem cell transplantation. Current standard of grading system is based on clinical symptoms in skin, liver and intestinal. However, it’s difficult to differ GVHD and its extent just by clinical manifestation. Here we retrospectively analyzed cell immune function in patients implemented allogeneic stem cell transplantation in Ningbo first Hospital from Jan 2013 to Jan 2018. Results the data are collected from 51 patients (mean age was 42; 45.1% women). The average NK cell percentage was 39.31% in severe GVHD (Grade III-IV), was 16.98% in mild GVHD (GradeI-II), while was 21.15% in No GVHD group. The statistical analysis showed difference among each grade. Further analysis was performed in Antithymocyte globulin (ATG) treated group and control group. We showed NK Cell percentage was sharply different in ATG treated group: 47.34% in severe GVHD, 11.98% in mild GVHD group, while 18.3% in no GVHD group. However, in control group, the average percentage of NK cells was 23.27% in severe GVHD, was 23.22%in mild GVHD group, while was 21.13% in no GVHD group. Conclusion The data supports that ATG can prevent GVHD by increasing NK cell percentage. The percentage of NK cell seemed to be a useful probe to evaluate the severity of GVHD in allogeneic stem cell transplantation patients using ATG in pretreatment.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Hematology, Ningbo First Hospital, Ningbo, 315010, China.
| | - Shujun Yang
- Laboratory of Stem Cell Transplantation, Ningbo First Hospital, Ningbo, 315010, China
| | - Yujing Zou
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC, 27710, USA
| | - Xiao Yan
- Department of Hematology, Ningbo First Hospital, Ningbo, 315010, China
| | - Hao Wu
- Department of Hematology, Ningbo First Hospital, Ningbo, 315010, China
| | - Miao Zhou
- Department of Hematology, Ningbo First Hospital, Ningbo, 315010, China
| | - Yong Cheng Sun
- Department of Hematology, Ningbo First Hospital, Ningbo, 315010, China
| | - Yi Zhang
- Laboratory of Stem Cell Transplantation, Ningbo First Hospital, Ningbo, 315010, China
| | - Huiling Zhu
- Department of Hematology, Ningbo First Hospital, Ningbo, 315010, China
| | - Kaihong Xu
- Department of Hematology, Ningbo First Hospital, Ningbo, 315010, China
| | - Yi Wang
- Department of Hematology, Ningbo First Hospital, Ningbo, 315010, China
| | - Li Xia Sheng
- Department of Hematology, Ningbo First Hospital, Ningbo, 315010, China
| | - Qitian Mu
- Laboratory of Stem Cell Transplantation, Ningbo First Hospital, Ningbo, 315010, China
| | - Liguang Sun
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China.
| | - Guifang Ouyang
- Department of Hematology, Ningbo First Hospital, Ningbo, 315010, China.
| |
Collapse
|
75
|
Marks DH, Naftulin JS, Penzi LR, Manatis-Lornell A, Yasuda MR, Chapman CM, Rao SR, Saavedra A, Senna MM. Histologic and clinical cross-sectional study of chronic hair loss in patients with cutaneous chronic graft-versus-host disease. J Am Acad Dermatol 2019; 81:1134-1141. [DOI: 10.1016/j.jaad.2019.03.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 02/15/2019] [Accepted: 03/08/2019] [Indexed: 12/21/2022]
|
76
|
Impact of low-dose anti-thymocyte globulin on immune reconstitution after allogeneic hematopoietic cell transplantation. Int J Hematol 2019; 111:120-130. [DOI: 10.1007/s12185-019-02756-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/30/2019] [Accepted: 09/30/2019] [Indexed: 01/13/2023]
|
77
|
Batorov EV, Pronkina NV, Tikhonova MA, Kryuchkova IV, Sergeevicheva VV, Sizikova SA, Ushakova GY, Aristova TA, Batorova DS, Shishkova IV, Gilevich AV, Shevela EY, Ostanin AA, Chernykh ER. Increased circulating CD3 + T cells are associated with early relapse following autologous hematopoietic stem cell transplantation in patients with classical Hodgkin lymphoma. Leuk Lymphoma 2019; 60:2488-2497. [PMID: 31609150 DOI: 10.1080/10428194.2019.1581934] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Non-malignant host immune cells are the main substrate in classical Hodgkin lymphoma (HL) microenvironment. Reconstitution of lymphocyte populations following the high-dose chemotherapy (HDC) with autologous hematopoietic stem cell transplantation (auto-HSCT) can support tumor growth in HL patients. We investigated recovery dynamics of circulating CD3+, CD4+, CD8+, CD16+/CD56+, CD19+, CD4+FOXP3+ lymphocytes following auto-HSCT in 79 HL patients and assessed relationship between these populations and the development of early relapse. Studied populations were not statistically significant between patients with high or standard/intermediate risk of relapse. CD3+ T cells at the time of engraftment were increased in patients with the early relapse of HL compared to non-relapsed patients (PU = 0.0028). Area under the curve was 0.76 (р = .0037). In logistic regression models, CD3+ T cell count was associated with early relapse/progression as a trend. These findings elucidate several interactions between early systemic T cell recovery and tumor progression following HDC with auto-HSCT.
Collapse
Affiliation(s)
- Egor V Batorov
- Laboratory of Cellular Immunotherapy, Research Institute of Fundamental and Clinical Immunology , Novosibirsk , Russia
| | - Natalia V Pronkina
- Laboratory of Clinical Immunology, Research Institute of Fundamental and Clinical Immunology , Novosibirsk , Russia
| | - Marina A Tikhonova
- Laboratory of Cellular Immunotherapy, Research Institute of Fundamental and Clinical Immunology , Novosibirsk , Russia
| | - Irina V Kryuchkova
- Department of Hematology and Bone Marrow Transplantation, Research Institute of Fundamental and Clinical Immunology , Novosibirsk , Russia
| | - Vera V Sergeevicheva
- Department of Hematology and Bone Marrow Transplantation, Research Institute of Fundamental and Clinical Immunology , Novosibirsk , Russia
| | - Svetlana A Sizikova
- Department of Hematology and Bone Marrow Transplantation, Research Institute of Fundamental and Clinical Immunology , Novosibirsk , Russia
| | - Galina Y Ushakova
- Department of Hematology and Bone Marrow Transplantation, Research Institute of Fundamental and Clinical Immunology , Novosibirsk , Russia
| | - Tatiana A Aristova
- Department of Hematology and Bone Marrow Transplantation, Research Institute of Fundamental and Clinical Immunology , Novosibirsk , Russia
| | - Dariya S Batorova
- Department of Hematology and Bone Marrow Transplantation, Research Institute of Fundamental and Clinical Immunology , Novosibirsk , Russia
| | - Irina V Shishkova
- Laboratory of Clinical Immunology, Research Institute of Fundamental and Clinical Immunology , Novosibirsk , Russia
| | - Andrey V Gilevich
- Intensive Care Unit, Research Institute of Fundamental and Clinical Immunology , Novosibirsk , Russia
| | - Ekaterina Y Shevela
- Laboratory of Cellular Immunotherapy, Research Institute of Fundamental and Clinical Immunology , Novosibirsk , Russia
| | - Alexander A Ostanin
- Laboratory of Cellular Immunotherapy, Research Institute of Fundamental and Clinical Immunology , Novosibirsk , Russia
| | - Elena R Chernykh
- Laboratory of Cellular Immunotherapy, Research Institute of Fundamental and Clinical Immunology , Novosibirsk , Russia
| |
Collapse
|
78
|
Gu G, Yang JZ, Zhang JQ, Sun LX. Regulatory T cells in allogeneic hematopoietic stem cell transplantation: From the lab to the clinic. Cell Immunol 2019; 346:103991. [PMID: 31607390 DOI: 10.1016/j.cellimm.2019.103991] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 09/19/2019] [Accepted: 10/01/2019] [Indexed: 12/14/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a curable strategy for the treatment of hematological malignancies and nonmalignant diseases. However, graft-versus-host disease (GVHD) and relapse are still two major causes of morbidity and mortality after allo-HSCT, and both restrict the improvement of transplant outcomes. Regulatory T cells (Tregs) has been successfully used in allo-SCT settings. In this review, we summarize recent advances in experimental studies that have evaluated the roles played by Tregs in the establishment of novel transplant modalities, the prevention of GVHD and the enhancement of immune reconstitution. We also discuss the application of Tregs in clinical to prevent acute GVHD, treat chronic GVHD, as well as enhance immune reconstitution and decrease leukemia relapse, all of which lead to improving transplant outcomes.
Collapse
Affiliation(s)
- Guang Gu
- Department of Rheumatology, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jian-Zhu Yang
- Department of Pathology, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jin-Qiao Zhang
- Department of Hematology, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Li-Xia Sun
- Department of Hematology, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
79
|
Nakamae H, Fujii K, Nanno S, Okamura H, Nakane T, Koh H, Nakashima Y, Nakamae M, Hirose A, Teshima T, Hino M. A prospective observational study of immune reconstitution following transplantation with post‐transplant reduced‐dose cyclophosphamide from
HLA
‐haploidentical donors. Transpl Int 2019; 32:1322-1332. [DOI: 10.1111/tri.13494] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 03/06/2019] [Accepted: 08/15/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Hirohisa Nakamae
- Hematology Graduate School of Medicine Osaka City University Osaka Japan
| | - Kazuki Fujii
- Department of Clinical Laboratory Osaka City University Hospital Osaka Japan
| | - Satoru Nanno
- Hematology Graduate School of Medicine Osaka City University Osaka Japan
| | - Hiroshi Okamura
- Hematology Graduate School of Medicine Osaka City University Osaka Japan
| | - Takahiko Nakane
- Hematology Graduate School of Medicine Osaka City University Osaka Japan
| | - Hideo Koh
- Hematology Graduate School of Medicine Osaka City University Osaka Japan
| | - Yasuhiro Nakashima
- Hematology Graduate School of Medicine Osaka City University Osaka Japan
| | - Mika Nakamae
- Hematology Graduate School of Medicine Osaka City University Osaka Japan
| | - Asao Hirose
- Hematology Graduate School of Medicine Osaka City University Osaka Japan
| | - Takanori Teshima
- Department of Hematology Faculty of Medicine Hokkaido University Sapporo Japan
| | - Masayuki Hino
- Hematology Graduate School of Medicine Osaka City University Osaka Japan
| |
Collapse
|
80
|
Saad A, Lamb L, Wang T, Hemmer MT, Spellman S, Couriel D, Alousi A, Pidala J, Abdel-Azim H, Agrawal V, Aljurf M, Beitinjaneh AM, Bhatt VR, Buchbinder D, Byrne M, Cahn JY, Cairo M, Castillo P, Chhabra S, Diaz MA, Farhan S, Floisand Y, Frangoul HA, Gadalla SM, Gajewski J, Gale RP, Gandhi M, Gergis U, Hamilton BK, Hematti P, Hildebrandt GC, Kamble RT, Kanate AS, Khandelwal P, Lazaryan A, MacMillan M, Marks DI, Martino R, Mehta PA, Nishihori T, Olsson RF, Patel SS, Qayed M, Rangarajan HG, Reshef R, Ringden O, Savani BN, Schouten HC, Schultz KR, Seo S, Shaffer BC, Solh M, Teshima T, Urbano-Ispizua A, Verdonck LF, Vij R, Waller EK, William B, Wirk B, Yared JA, Yu LC, Arora M, Hashmi S. Impact of T Cell Dose on Outcome of T Cell-Replete HLA-Matched Allogeneic Peripheral Blood Stem Cell Transplantation. Biol Blood Marrow Transplant 2019; 25:1875-1883. [PMID: 31085303 PMCID: PMC7071947 DOI: 10.1016/j.bbmt.2019.05.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/02/2019] [Accepted: 05/03/2019] [Indexed: 01/24/2023]
Abstract
Data on whether the T cell dose of allogeneic peripheral blood stem cell (PBSC) products influences transplantation outcomes are conflicting. Using the Center for International Blood and Marrow Transplant Research database, we identified 2736 adult patients who underwent first allogeneic PBSC transplantation for acute leukemia or myelodysplastic syndrome between 2008 and 2014 using an HLA-matched sibling donor (MSD) or an 8/8-matched unrelated donor (MUD). We excluded ex vivo and in vivo T cell-depleted transplantations. Correlative analysis was performed between CD3+ T cell dose and the risk of graft-versus-host-disease (GVHD), relapse, nonrelapse mortality (NRM), disease-free survival (DFS), and overall survival (OS). Using maximum likelihood estimation, we identified CD3+ T cell dose cutoff that separated the risk of acute GVHD (aGVHD) grade II-IV in both the MSD and MUD groups. A CD3+ T cell dose cutoff of 14 × 107 cells/kg identified MSD/low CD3+ (n = 223) and MSD/high CD3+ (n = 1214), and a dose of 15 × 107 cells/kg identified MUD/low CD3+ (n = 197) and MUD/high CD3+ (n = 1102). On univariate analysis, the MSD/high CD3+ group had a higher cumulative incidence of day +100 aGVHD grade II-IV compared with the MSD/low CD3+ group (33% versus 25%; P = .009). There were no differences between the 2 groups in engraftment rate, risk of aGVHD grade III-IV or chronic GVHD (cGVHD), NRM, relapse, DFS, or OS. The MUD/high CD3+ group had a higher cumulative incidence of day +100 aGVHD grade II-IV compared with the MUD/low CD3+ group (49% versus 41%; P = .04). There were no differences between the 2 groups in engraftment rate, risk of severe aGVHD or cGVHD, NRM, relapse, DFS, or OS. Multivariate analysis of the MSD and MUD groups failed to show an association between CD3+ T cell dose and the risk of either aGVHD grade II-IV (P = .10 and .07, respectively) or cGVHD (P = .80 and .30, respectively). Subanalysis of CD4+ T cells, CD8+ T cells, and CD4+/CD8+ ratio failed to identify cutoff values predictive of transplantation outcomes; however, using the log-rank test, the sample size was suboptimal for identifying a difference at this cutoff cell dose. In this registry study, the CD3+ T cell dose of PBSC products did not influence the risk of aGVHD or cGVHD or other transplantation outcomes when using an MSD or an 8/8-matched MUD. Subset analyses of CD4+ and CD8+ T cell doses were not possible given our small sample size.
Collapse
Affiliation(s)
- Ayman Saad
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Lawrence Lamb
- University of Alabama at Birmingham, Birmingham, Alabama
| | - Tao Wang
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, Wisconsin; Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Michael T Hemmer
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Stephen Spellman
- Center for International Blood and Marrow Transplant Research, National Marrow Donor Program/Be the Match, Minneapolis, Minnesota
| | - Daniel Couriel
- Utah Blood and Marrow Transplant Program, Salt Lake City, Utah
| | - Amin Alousi
- Department of Stem Cell Transplantation, Division of Cancer Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Joseph Pidala
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Hisham Abdel-Azim
- Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital of Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California
| | - Vaibhav Agrawal
- Division of Hematology-Oncology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Mahmoud Aljurf
- Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | | | - Vijaya Raj Bhatt
- The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - David Buchbinder
- Division of Pediatric Hematology, Children's Hospital of Orange County, Orange, California
| | - Michael Byrne
- Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jean-Yves Cahn
- Department of Hematology, CHU Grenoble Alpes, Grenoble, France
| | - Mitchell Cairo
- Division of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Department of Pediatrics, New York Medical College, New York, New York
| | - Paul Castillo
- UF Health Shands Children's Hospital, Gainesville, Florida
| | - Saurabh Chhabra
- Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Miguel Angel Diaz
- Department of Hematology/Oncology, Hospital Infantil Universitario Nino Jesus, Madrid, Spain
| | - Shatha Farhan
- Henry Ford Hospital Bone Marrow Transplant Program, Detroit, Michigan
| | | | - Hadar A Frangoul
- Children's Hospital at TriStar Centennial and Sarah Cannon Research Institute, Nashville, Tennessee
| | - Shahinaz M Gadalla
- Division of Cancer Epidemiology & Genetics, Clinical Genetics Branch, National Cancer Institute, Rockville, Maryland
| | | | - Robert Peter Gale
- Hematology Research Center, Division of Experimental Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| | - Manish Gandhi
- Division of Transfusion Medicine, Mayo Clinic, Rochester, Minnesota
| | - Usama Gergis
- Hematologic Malignancies & Bone Marrow Transplant, Department of Medical Oncology, New York Presbyterian Hospital/Weill Cornell Medical Center, New York, New York
| | - Betty Ky Hamilton
- Blood & Marrow Transplant Program, Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio
| | - Peiman Hematti
- Division of Hematology/Oncology/Bone Marrow Transplantation, Department of Medicine, University of Wisconsin Hospital and Clinics, Madison, Wisconsin
| | | | - Rammurti T Kamble
- Division of Hematology and Oncology, Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Abraham S Kanate
- Osborn Hematopoietic Malignancy and Transplantation Program, West Virginia University, Morgantown, West Virginia
| | - Pooja Khandelwal
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Aleksandr Lazaryan
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Margaret MacMillan
- University of Minnesota Blood and Marrow Transplant Program, Pediatrics, Minneapolis, Minnesota
| | - David I Marks
- Adult Bone Marrow Transplant, University Hospitals Bristol NHS Trust, Bristol, United Kingdom
| | - Rodrigo Martino
- Division of Clinical Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Parinda A Mehta
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Taiga Nishihori
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Richard F Olsson
- Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden; Centre for Clinical Research Sormland, Uppsala University, Uppsala, Sweden
| | - Sagar S Patel
- Blood and Marrow Transplant Program, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Muna Qayed
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Hemalatha G Rangarajan
- Department of Pediatric Hematology, Oncology, Blood and Marrow Transplantation, Nationwide Children's Hospital, Columbus, Ohio
| | - Ran Reshef
- Blood and Marrow Transplantation Program and Columbia Center for Translational Immunobiology, Columbia University Medical Center, New York, New York
| | - Olle Ringden
- Translational Cell Therapy Research, Karolinska Institute, Stockholm, Sweden
| | - Bipin N Savani
- Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Harry C Schouten
- Department of Hematology, Academische Ziekenhuis, Maastricht, Netherlands
| | - Kirk R Schultz
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplant, British Columbia's Children's Hospital, The University of British Columbia, Vancouver, Britich Columbia, Canada
| | - Sachiko Seo
- Department of Hematology and Oncology, Dokkyo Medical University, Tochigi, Japan
| | | | - Melhem Solh
- The Blood and Marrow Transplant Group of Georgia, Northside Hospital, Atlanta, Georgia
| | | | - Alvaro Urbano-Ispizua
- Department of Hematology, Hospital Clinic, University of Barcelona, IDIBAPS, and Josep Carreras Institute of Research, Barcelona, Spain
| | - Leo F Verdonck
- Department of Hematology/Oncology, Isala Clinic, Zwolle, The Netherlands
| | - Ravi Vij
- Division of Hematology and Oncology, Washington University School of Medicine, St Louis, Missouri
| | - Edmund K Waller
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Basem William
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Baldeep Wirk
- Division of Bone Marrow Transplant, Seattle Cancer Care Alliance, Seattle, Washington
| | - Jean A Yared
- Blood & Marrow Transplantation Program, Division of Hematology/Oncology, Department of Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland
| | - Lolie C Yu
- Division of Hematology/Oncology and HSCT, Center for Cancer and Blood Disorders, Children's Hospital/Louisiana State University Medical Center, New Orleans, Louisiana
| | - Mukta Arora
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota Medical Center, Minneapolis, Minnesota.
| | - Shahrukh Hashmi
- Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia; Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
81
|
Wang F, Zhao S, Gu Z, Zhao X, Yang N, Guan L, Liu T, Wang L, Fang S, Zhu C, Luo L, Li M, Wang L, Gao C. S1PR5 regulates NK cell responses in preventing graft-versus-host disease while preserving graft-versus-tumour activity in a murine allogeneic haematopoietic stem cell transplantation model. Hematol Oncol 2019; 38:89-102. [PMID: 31465552 DOI: 10.1002/hon.2669] [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: 07/04/2019] [Revised: 08/22/2019] [Accepted: 08/23/2019] [Indexed: 11/09/2022]
Abstract
Graft-versus-host disease (GVHD) remains a major complication following allogeneic haematopoietic stem cell transplantation (allo-HSCT) leading to high transplant-related mortality. Natural killer (NK) cells have been found to mitigate GVHD without attenuating the graft-versus-tumour (GVT) activity in the murine model of haematopoietic stem cell transplantation. Sphingosine-1-phosphate receptor 5 (S1PR5) is a very important chemokine receptor on NK cells that governs NK cell distribution in vivo and trafficking at lesion sites. Our preliminary studies showed that the incidence of GVHD was negatively correlated with S1PR5 expression in the NK cells of patients after allo-HSCT. In the present study, we found that S1PR5 deficiency in murine NK cells blocked the migration of NK cells from the bone marrow to the GVHD target organs and attenuated the inhibitory effects on the alloreactive T cells, especially CD3+ CD8+ T cells, which may be the reason why the loss of S1PR5 in NK cells could aggravate GVHD in recipient mice. Furthermore, we also demonstrated that the absence of S1PR5 expression in NK cells did not interfere with the antitumour effects of NK cells and T cells in vivo. Taken together, our data indicate that S1PR5 plays an essential role in balancing GVHD and GVT activity.
Collapse
Affiliation(s)
- Feiyan Wang
- Medical School, Nankai University, Tianjin, China.,Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Shasha Zhao
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,Department of Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Zhenyang Gu
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Xiaoli Zhao
- Department of Hematology, Chinese Academy of Medical Sciences, Tianjin, China
| | - Nan Yang
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Lixun Guan
- Department of Hematology, Hainan Branch, PLA General Hospital, Hainan, China
| | - Tong Liu
- Inpatient Department, 66242 Army Hospital, Xilin Gol, China
| | - Li Wang
- Department of Hematology and Oncology, Laoshan Branch, Chinese PLA 401 Hospital, Qingdao, China
| | - Shu Fang
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Chengying Zhu
- Medical School, Nankai University, Tianjin, China.,Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Lan Luo
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Meng Li
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Lili Wang
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Chunji Gao
- Medical School, Nankai University, Tianjin, China.,Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
82
|
Zhang P, Tey SK. Adoptive T Cell Therapy Following Haploidentical Hematopoietic Stem Cell Transplantation. Front Immunol 2019; 10:1854. [PMID: 31447852 PMCID: PMC6691120 DOI: 10.3389/fimmu.2019.01854] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 07/23/2019] [Indexed: 12/24/2022] Open
Abstract
Delayed immune reconstitution and the consequently high rates of leukemia relapse and infectious complications are the main limitations of haploidentical hematopoietic stem cell transplantation. Donor T cell addback can accelerate immune reconstitution but the therapeutic window between graft-vs.-host disease and protective immunity is very narrow in the haploidentical transplant setting. Hence, strategies to improve the safety and efficacy of adoptive T cell transfer are particularly relevant in this setting. Adoptive T cell transfer strategies in haploidentical transplantation include the use of antigen-specific T cells, allodepletion and alloanergy induction, immune modulation by the co-infusion of regulatory cell populations, and the use of safety switch gene-modified T cells. Whilst common principles apply, there are features that are unique to haploidentical transplantation, where HLA-mismatching directly impacts on immune reconstitution, and shared vs. non-shared HLA-allele can be an important consideration in antigen-specific T cell therapy. This review will also present an update on safety switch gene-modified T cells, which can be conditionally deleted in the event of severe graft- vs.-host disease or other adverse events. Herpes Virus Simplex Thymidine Kinase (HSVtk) and inducible caspase-9 (iCasp9) are safety switches that have undergone multicenter studies in haploidentical transplantation with encouraging results. These gene-modified cells, which are trackable long-term, have also provided important insights on the fate of adoptively transferred T cells. In this review, we will discuss the biology of post-transplant T cell immune reconstitution and the impact of HLA-mismatching, and the different cellular therapy strategies that can help accelerate T cell immune reconstitution after haploidentical transplantation.
Collapse
Affiliation(s)
- Ping Zhang
- Clinical Translational Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Siok-Keen Tey
- Clinical Translational Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia.,Department of Haematology and Bone Marrow Transplantation, Royal Brisbane and Women's Hospital, Herston, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
83
|
Effect of Sirolimus on Immune Reconstitution Following Myeloablative Allogeneic Stem Cell Transplantation: An Ancillary Analysis of a Randomized Controlled Trial Comparing Tacrolimus/Sirolimus and Tacrolimus/Methotrexate (Blood and Marrow Transplant Clinical Trials Network/BMT CTN 0402). Biol Blood Marrow Transplant 2019; 25:2143-2151. [PMID: 31271885 DOI: 10.1016/j.bbmt.2019.06.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 06/26/2019] [Accepted: 06/26/2019] [Indexed: 11/21/2022]
Abstract
Although allogeneic hematopoietic cell transplantation (HCT) is a potentially curative therapy for hematologic neoplasms, one of its limiting toxicities continues to be graft-versus-host disease, both acute (aGVHD) and chronic (cGVHD). Sirolimus is a mammalian target of rapamycin inhibitor that has proven effective in GVHD prophylaxis in combination with a calcineurin inhibitor, such as tacrolimus. The impact of sirolimus on immune reconstitution has not been comprehensively investigated in vivo thus far, however. Here we present an ancillary analysis of the randomized study BMT-CTN 0402 that examined the effect of sirolimus on immune subsets post-transplantation. We further examine the association between different lymphocyte subsets and outcomes post-transplantation in each arm. BMT-CTN 0402 was a randomized trial (n = 304) comparing 2 GVHD prophylaxis regimens, tacrolimus/sirolimus (Tac/Sir) and tacrolimus/methotrexate (Tac/MTX), in patients with acute myelogenous leukemia, acute lymphoblastic leukemia, or myelodysplastic syndrome undergoing myeloablative HLA-matched HCT. There were no differences in 114-day GVHD-free survival (primary endpoint), aGVHD, cGVHD, relapse, or overall survival (OS) between the 2 arms. Of the 304 patients, 264 had available samples for the current immune reconstitution analysis. Blood samples were collected at 1, 3, 6, 12, and 24 months post-HCT. Multiparameter flow cytometry was performed at the project laboratory (Esoterix Clinical Trials Services) in a blinded fashion, and results for the 2 arms were compared. Multivariable Cox regression models, treating each phenotypic parameter as a time-dependent variable, were constructed to study the impact of reconstitution on clinical outcomes. There were no significant differences in patient and transplantation characteristics between the Tac/Sir and Tac/MTX arms in this analysis. Absolute lymphocyte count and CD3+ cell, CD4+ cell, and conventional T cell (Tcon) counts were significantly decreased in the Tac/Sir arm for up to 3 months post-HCT, whereas CD8+ cells recovered even more slowly (up to 6 months) in this arm. Interestingly, there was no clear difference in the absolute number of regulatory T cells (Tregs, defined as CD4+CD25+ cells) between the 2 arms at any point post-HCT; however, the Treg:Tcon ratio was significantly greater in the Tac/Sir arm in the first 3 months after HCT. B lymphocyte recovery was significantly compromised in the Tac/Sir arm from 1 month to 6 months after HCT, whereas natural killer cell reconstitution was not affected in the Tac/Sir arm. In the outcomes analysis, higher numbers of CD3+ cells, CD4+ cells, CD8+ cells, and Tregs were associated with better OS. Neither Treg numbers nor the Treg:Tcon ratio was correlated with GVHD. Our findings indicate that Tac/Sir has a more profound T cell suppressive effect than the combination of Tac/MTX in the early post-transplantation period, and particularly compromises the recovery of CD8+ T cells, which have been implicated in aGVHD. Sirolimus used in vivo with tacrolimus does not appear to result in increased absolute numbers of Tregs, but might have a beneficial effect on the Treg:Tcon balance in the first 3 months after transplantation. Nonetheless, no differences in aGVHD or cGVHD between the 2 arms were observed in the parent randomized trial. Calcineurin-inhibitor free, sirolimus-containing GVHD prophylaxis strategies, incorporating other novel agents, should be investigated further to maximize the potential favorable effect of sirolimus on Treg:Tcon balance in the post-transplantation immune repertoire. Sirolimus significantly compromises B cell recovery in the first 6 months post-HCT, with potential complex effects on cGVHD that merit further study.
Collapse
|
84
|
Copsel S, Wolf D, Komanduri KV, Levy RB. The promise of CD4 +FoxP3 + regulatory T-cell manipulation in vivo: applications for allogeneic hematopoietic stem cell transplantation. Haematologica 2019; 104:1309-1321. [PMID: 31221786 PMCID: PMC6601084 DOI: 10.3324/haematol.2018.198838] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/07/2019] [Indexed: 12/12/2022] Open
Abstract
CD4+FoxP3+ regulatory T cells (Tregs) are a non-redundant population critical for the maintenance of self-tolerance. Over the past decade, the use of these cells for therapeutic purposes in transplantation and autoimmune disease has emerged based on their capacity to inhibit immune activation. Basic science discoveries have led to identifying key receptors on Tregs that can regulate their proliferation and function. Notably, the understanding that IL-2 signaling is crucial for Treg homeostasis promoted the hypothesis that in vivo IL-2 treatment could provide a strategy to control the compartment. The use of low-dose IL-2 in vivo was shown to selectively expand Tregs versus other immune cells. Interestingly, a number of other Treg cell surface proteins, including CD28, CD45, IL-33R and TNFRSF members, have been identified which can also induce activation and proliferation of this population. Pre-clinical studies have exploited these observations to prevent and treat mice developing autoimmune diseases and graft-versus-host disease post-allogeneic hematopoietic stem cell transplantation. These findings support the development of translational strategies to expand Tregs in patients. Excitingly, the use of low-dose IL-2 for patients suffering from graft-versus-host disease and autoimmune disease has demonstrated increased Treg levels together with beneficial outcomes. To date, promising pre-clinical and clinical studies have directly targeted Tregs and clearly established the ability to increase their levels and augment their function in vivo. Here we review the evolving field of in vivo Treg manipulation and its application to allogeneic hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
| | | | - Krishna V Komanduri
- Department of Microbiology and Immunology.,Sylvester Comprehensive Cancer Center.,Division of Transplantation and Cellular Therapy, Department of Medicine
| | - Robert B Levy
- Department of Microbiology and Immunology .,Division of Transplantation and Cellular Therapy, Department of Medicine.,Department of Ophthalmology, Miller School of Medicine, University of Miami, FL, USA
| |
Collapse
|
85
|
Li M, Lu C, Zhu H, Kang X, Wang F, Shao L, Lu X, Chen W, Xia X. Cenicriviroc ameliorates the severity of graft-versus-host disease through inhibition of CCR5 in a rat model of liver transplantation. Am J Transl Res 2019; 11:3438-3449. [PMID: 31312356 PMCID: PMC6614659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 05/30/2019] [Indexed: 06/10/2023]
Abstract
Acute graft-versus-host disease (aGVHD) is one of the major complications after liver transplantation (LTx), which is induced by over-activation of T helper lymphocytes. Cenicriviroc (CVC) exerts its anti-inflammatory effect through inhibition of C-C chemokine receptor 5 (CCR5). However, whether CVC ameliorates aGVHD after liver transplantation remains unknown. In the present study, a rat aGVHD liver transplantation model (LTx-aGVHD) was constructed. CVC was intravenously injected from day 7 to day 14 after LTx. Liver and intestine samples were harvested to evaluate GVHD severity. Peripheral blood mononuclear cells (PBMCs) were collected and CCR5 antibodies were prepared to further explore the molecular mechanism in vitro. CVC significantly decreased the severity of GVHD associated skin and intestine injury. Quality of life of the LTx-GVHD rats was improved after CVC treatment. Flow cytometry further confirmed diminished peripheral donor-derived Th cells after CVC treatment. Molecularly, CVC treatment showed similar anti-inflammatory effects to CCR5 antibody injection. The level of CCR5, C-C motif chemokine ligand 5 (CCL5), and pro-inflammatory cytokines in the liver and intestines were inhibited after CVC treatment. Thus, CVC deactivated Th lymphocytes and decreased the severity of LTx-aGVHD through inhibition of CCR5.
Collapse
Affiliation(s)
- Minhuan Li
- Center of Pathology and Clinical Laboratory, Sir Run Run Hospital, Nanjing Medical UniversityNanjing 211100, Jiangsu Province, China
| | - Chenglin Lu
- Department of General Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing 210008, Jiangsu Province, China
| | - Hao Zhu
- Department of Gastroenterologz, The Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing 210008, Jiangsu Province, China
| | - Xing Kang
- Department of General Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing 210008, Jiangsu Province, China
| | - Feng Wang
- Department of General Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing 210008, Jiangsu Province, China
| | - Lihua Shao
- Department of General Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing 210008, Jiangsu Province, China
| | - Xiaofeng Lu
- Department of General Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing 210008, Jiangsu Province, China
| | - Wei Chen
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang ProvinceHangzhou 310012, Zhejiang Province, China
| | - Xuefeng Xia
- Department of General Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing 210008, Jiangsu Province, China
| |
Collapse
|
86
|
Törlén J, Gaballa A, Remberger M, Mörk LM, Sundberg B, Mattsson J, Uhlin M. Effect of Graft-versus-Host Disease Prophylaxis Regimens on T and B Cell Reconstitution after Allogeneic Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant 2019; 25:1260-1268. [DOI: 10.1016/j.bbmt.2019.01.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 01/17/2019] [Indexed: 01/06/2023]
|
87
|
Yang S, Sheng X, Xiang D, Wei X, Chen T, Yang Z, Zhang Y. CD150 highTreg cells may attenuate graft versus host disease and intestinal cell apoptosis after hematopoietic stem cell transplantation. Am J Transl Res 2019; 11:1299-1310. [PMID: 30972163 PMCID: PMC6456532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 01/29/2019] [Indexed: 06/09/2023]
Abstract
Combined transplantation of regulatory T cells (Treg cells) may significantly attenuate graft versus host disease (GVHD) after hematopoietic stem cell transplantation (HSCT). Recent studies indicated that CD150+Treg cells could secret adenosine to maintain the quiescent status of HSCs. However, whether it is attributable to the attenuation of GVHD after HSCT is still unclear. In vitro studies revealed that CD150+Treg cells induced immune tolerance was comparable to that induced by CD150-Treg cells, but CD150+Treg cells can secret more adenosine, increase P-AMPK expression and regulate energy metabolism to induce the proliferation of HSC proliferation and inhibit their differentiation into dendritic cells. In this study, GVHD animal model was established, and combined transplantation of Treg cells and HSCs was performed. Results showed the survival time was significantly prolonged, the proliferation rate of HSCs increased significantly and the proportion of undifferentiated HSCs elevated significantly after CD150+Treg transplantation as compared to CD150-Treg transplantation. Immunohistochemistry revealed CD150+Treg cells could secret adenosine, activate AMPK expression and inhibit intestinal cell apoptosis and inflammation after HSCT. Taken together, this study indicates CD150+Treg cells can regulate energy metabolism to attenuate GVHD and intestinal cell apoptosis after HSCT.
Collapse
Affiliation(s)
- Sainan Yang
- Department of Hematology, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital) Chongqing 401120, China
| | - Xiaomin Sheng
- Department of Hematology, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital) Chongqing 401120, China
| | - Dan Xiang
- Department of Hematology, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital) Chongqing 401120, China
| | - Xia Wei
- Department of Hematology, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital) Chongqing 401120, China
| | - Tingting Chen
- Department of Hematology, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital) Chongqing 401120, China
| | - Zailin Yang
- Department of Hematology, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital) Chongqing 401120, China
| | - Yong Zhang
- Department of Hematology, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital) Chongqing 401120, China
| |
Collapse
|
88
|
Soares MV, Azevedo RI, Ferreira IA, Bucar S, Ribeiro AC, Vieira A, Pereira PNG, Ribeiro RM, Ligeiro D, Alho AC, Soares AS, Camacho N, Martins C, Lourenço F, Moreno R, Ritz J, Lacerda JF. Naive and Stem Cell Memory T Cell Subset Recovery Reveals Opposing Reconstitution Patterns in CD4 and CD8 T Cells in Chronic Graft vs. Host Disease. Front Immunol 2019; 10:334. [PMID: 30894856 PMCID: PMC6414429 DOI: 10.3389/fimmu.2019.00334] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/08/2019] [Indexed: 01/05/2023] Open
Abstract
The success of allogeneic hematopoietic stem cell transplantation (allo-HSCT) in the treatment of hematological malignancies remains hampered by life-threatening chronic graft vs. host disease (cGVHD). Although multifactorial in nature, cGVHD has been associated with imbalances between effector and regulatory T cells (Treg). To further elucidate this issue, we performed a prospective analysis of patients undergoing unrelated donor allo-HSCT after a reduced intensity conditioning (RIC) regimen containing anti-thymocyte globulin (ATG) and the same GVHD prophylaxis, at a single institution. We studied T cell subset homeostasis over a 24-month follow-up after HSCT in a comparative analysis of patients with and without cGVHD. We also quantified naive and memory T cell subsets, proliferation and expression of the apoptosis-related proteins Bcl-2 and CD95. Finally, we assessed thymic function by T cell receptor excision circle (TREC) quantification and T cell receptor (TCR) diversity by TCRVβ spectratyping. While the total number of conventional CD4 (Tcon) and CD8 T cells was similar between patient groups, Treg were decreased in cGVHD patients. Interestingly, we also observed divergent patterns of Naive and Stem Cell Memory (SCM) subset recovery in Treg and Tcon compared to CD8. Patients with cGVHD showed impaired recovery of Naive and SCM Tcon and Treg, but significantly increased frequencies and absolute numbers of Naive and SCM were observed in the CD8 pool. Markedly increased EMRA CD8 T cells were also noted in cGVHD. Taken together, these results suggest that Naive, SCM and EMRA CD8 play a role in the emergence of cGHVD. Reduced Naive and recent thymic emigrant Tcon and Treg in cGVHD was likely due to impaired thymic output, as it was accompanied by decreased CD4 TREC and TCR diversity. On the other hand, CD8 TCR diversity was similar between patient groups. Furthermore, no correlation was observed between CD8 TREC content and Naive CD8 numbers, suggesting limited thymic production of Naive CD8 T cells in patients after transplant, especially in those developing cGVHD. The mechanisms behind the opposing patterns of CD4 and CD8 subset cell recovery in cGVHD remain elusive, but may be linked to thymic damage associated with the conditioning regimen and/or acute GVHD.
Collapse
Affiliation(s)
- Maria V Soares
- JLacerda Lab, Hematology and Transplantation Immunology, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Rita I Azevedo
- JLacerda Lab, Hematology and Transplantation Immunology, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Inês A Ferreira
- JLacerda Lab, Hematology and Transplantation Immunology, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Sara Bucar
- JLacerda Lab, Hematology and Transplantation Immunology, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Ana C Ribeiro
- JLacerda Lab, Hematology and Transplantation Immunology, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Ana Vieira
- Unidade de Citometria de Fluxo, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Paulo N G Pereira
- JLacerda Lab, Hematology and Transplantation Immunology, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Ruy M Ribeiro
- Laboratório de Biomatemática, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Dario Ligeiro
- Lisbon Centre for Blood and Transplantation, Instituto Português do Sangue e Transplantação, IP, Lisbon, Portugal
| | - Ana C Alho
- JLacerda Lab, Hematology and Transplantation Immunology, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal.,Serviço de Hematologia e Transplantação de Medula, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Lisbon, Portugal
| | - António S Soares
- JLacerda Lab, Hematology and Transplantation Immunology, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Nádia Camacho
- Serviço de Hematologia e Transplantação de Medula, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Lisbon, Portugal
| | - Carlos Martins
- Serviço de Hematologia e Transplantação de Medula, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Lisbon, Portugal
| | - Fernanda Lourenço
- Serviço de Hematologia e Transplantação de Medula, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Lisbon, Portugal
| | - Raul Moreno
- Serviço de Hematologia e Transplantação de Medula, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Lisbon, Portugal
| | - Jerome Ritz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - João F Lacerda
- JLacerda Lab, Hematology and Transplantation Immunology, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal.,Serviço de Hematologia e Transplantação de Medula, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Lisbon, Portugal
| |
Collapse
|
89
|
Paczesny S, Metzger J. Clinical Proteomics for Post-Hematopoeitic Stem Cell Transplantation Outcomes. Proteomics Clin Appl 2019; 13:e1800145. [PMID: 30307119 PMCID: PMC6440827 DOI: 10.1002/prca.201800145] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 09/28/2018] [Indexed: 12/20/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is the most effective form of tumor immunotherapy available to date. However, while HSCT can induce beneficial graft-versus-leukemia (GVL) effect, the adverse effect of graft-versus-host disease (GVHD), which is closely linked to GVL, is the major source of morbidity and mortality following HSCT. Until recently, available diagnostic and staging tools frequently fail to identify those at higher risk of disease progression or death. Furthermore, there are shortcomings in the prediction of the need for therapeutic interventions or the response rates to different forms of therapy. The past decade has been characterized by an explosive evolution of proteomics technologies, largely due to important advances in high-throughput MS instruments and bioinformatics. Building on these opportunities, blood biomarkers have been identified and validated both as promising diagnostic tools, prognostic tools that risk-stratify patients before future occurrence of GVHD and as predictive tools for responsiveness to GVHD therapy and non-relapse mortality. These biomarkers might facilitate timely and selective therapeutic intervention. This review summarizes current information on clinical proteomics for GVHD as well as other complications following HSCT. Finally, it proposes future directions for the translation of clinical proteomics to discovery of new potential therapeutic targets to the development of drugs.
Collapse
Affiliation(s)
- Sophie Paczesny
- Department of Pediatrics, Department of Microbiology Immunology, and Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | |
Collapse
|
90
|
Trovillion EM, Gloude NJ, Anderson EJ, Morris GP. Relationship of post-transplant thymopoiesis with CD4 +FoxP3 + regulatory T cell recovery associated with freedom from chronic graft versus host disease. Bone Marrow Transplant 2018; 54:917-920. [PMID: 30413809 PMCID: PMC6509012 DOI: 10.1038/s41409-018-0394-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 10/27/2018] [Accepted: 10/30/2018] [Indexed: 02/07/2023]
Affiliation(s)
- Erin M Trovillion
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Nicholas J Gloude
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Eric J Anderson
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Gerald P Morris
- Department of Pathology, University of California San Diego, La Jolla, California, USA.
| |
Collapse
|
91
|
Groth C, van Groningen LFJ, Matos TR, Bremmers ME, Preijers FWMB, Dolstra H, Reicherts C, Schaap NPM, van Hooren EHG, IntHout J, Masereeuw R, Netea MG, Levine JE, Morales G, Ferrara JL, Blijlevens NMA, van Oosterhout YVJM, Stelljes M, van der Velden WJFM. Phase I/II Trial of a Combination of Anti-CD3/CD7 Immunotoxins for Steroid-Refractory Acute Graft-versus-Host Disease. Biol Blood Marrow Transplant 2018; 25:712-719. [PMID: 30399420 DOI: 10.1016/j.bbmt.2018.10.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 10/25/2018] [Indexed: 01/01/2023]
Abstract
Effective therapies for treating patients with steroid-refractory acute graft-versus-host-disease (SR-aGVHD), particularly strategies that reduce the duration of immunosuppression following remission, are urgently needed. The investigated immunotoxin combination consists of a mixture of anti-CD3 and anti-CD7 antibodies separately conjugated to recombinant ricin A (CD3/CD7-IT), which induces in vivo depletion of T cells and natural killer (NK) cells and suppresses T cell receptor activation. We conducted a phase I/II trial to examine the safety and efficacy of CD3/CD7-IT in 20 patients with SR-aGVHD; 17 of these patients (85%) had severe SR-aGVHD, and all 20 patients had visceral organ involvement, including 18 (90%) with gastrointestinal (GI) involvement and 5 (25%) with liver involvement. A validated 2-biomarker algorithm classified the majority of patients (11 of 20) as high risk. On day 28 after the start of CD3/CD7-IT therapy, the overall response rate was 60% (12 of 20), with 10 patients (50%) achieving a complete response. The 6-month overall survival rate was 60% (12 of 20), including 64% (7 of 11) classified as high risk by biomarkers. The 1-week course of treatment with CD3/CD7-IT caused profound but transient depletion of T cells and NK cells, followed by rapid recovery of the immune system with a diverse TCR Vβ repertoire, and preservation of Epstein-Barr virus- and cytomegalovirus-specific T cell clones. Furthermore, our results indicate that CD3/CD7-IT appeared to be safe and well tolerated, with a relatively low prevalence of manageable and reversible adverse events, primarily worsening of hypoalbuminemia, microangiopathy, and thrombocytopenia. These encouraging results suggest that CD3/CD7-IT may improve patient outcomes in patients with SR-aGVHD.
Collapse
Affiliation(s)
- Christoph Groth
- Department of Medicine A/Hematology and Oncology, University Hospital of Muenster, Muenster, Germany
| | - Lenneke F J van Groningen
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands; Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tiago R Matos
- Department of Dermatology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Manita E Bremmers
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frank W M B Preijers
- Department of Laboratory Medicine, Laboratory for Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Harry Dolstra
- Department of Laboratory Medicine, Laboratory for Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Christian Reicherts
- Department of Medicine A/Hematology and Oncology, University Hospital of Muenster, Muenster, Germany
| | - Nicolaas P M Schaap
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands; Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Joanna IntHout
- Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands; Section of Biostatistics, Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - John E Levine
- Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai Hospital, New York, NY
| | - George Morales
- Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai Hospital, New York, NY
| | - James L Ferrara
- Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai Hospital, New York, NY
| | - Nicole M A Blijlevens
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands; Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Matthias Stelljes
- Department of Medicine A/Hematology and Oncology, University Hospital of Muenster, Muenster, Germany
| | - Walter J F M van der Velden
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands; Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
92
|
Wang L, Ni M, Hückelhoven-Krauss A, Sellner L, Hoffmann JM, Neuber B, Luft T, Hegenbart U, Schönland S, Kleist C, Sill M, Chen BA, Wuchter P, Eckstein V, Krüger W, Hilgendorf I, Yerushalmi R, Nagler A, Müller-Tidow C, Ho AD, Dreger P, Schmitt M, Schmitt A. Modulation of B Cells and Homing Marker on NK Cells Through Extracorporeal Photopheresis in Patients With Steroid-Refractory/Resistant Graft-Vs.-Host Disease Without Hampering Anti-viral/Anti-leukemic Effects. Front Immunol 2018; 9:2207. [PMID: 30349527 PMCID: PMC6186805 DOI: 10.3389/fimmu.2018.02207] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/05/2018] [Indexed: 11/13/2022] Open
Abstract
Graft-vs.-host disease (GvHD), a severe complication of allogeneic hematopoietic stem cell transplantation, significantly affects the post-transplant morbidity and mortality. Systemic steroids remain the gold standard for the initial management of GvHD. However, up to 60% of patients will not sufficiently respond to steroids. Extracorporeal photopheresis (ECP), a cell-based immunotherapy, has shown good clinical results in such steroid-refractory/resistant GvHD patients. Given its immunomodulatory, but not global immunosuppressive and steroid-sparing capacity, ECP constitutes an attractive option. In the case of GvHD, the balance of immune cells is destroyed: effector cells are not any longer efficiently controlled by regulatory cells. ECP therapy may restore this balance. However, the precise mechanism and the impact of ECP on anti-viral/anti-leukemic function remain unclear. In this study, 839 ECP treatments were performed on patients with acute GvHD (aGvHD) and chronic GvHD (cGvHD). A comprehensive analysis of effector and regulatory cells in patients under ECP therapy included multi-parametric flow cytometry and tetramer staining, LuminexTM-based cytokine, interferon-γ enzyme-linked immunospot, and chromium-51 release assays. Gene profiling of myeloid-derived suppressor cells (MDSCs) was performed by microarray analysis. Immunologically, modulations of effector and regulatory cells as well as proinflammatory cytokines were observed under ECP treatment: (1) GvHD-relevant cell subsets like CD62L+ NK cells and newly defined CD19hiCD20hi B cells were modulated, but (2) quantity and quality of anti-viral/anti-leukemic effector cells were preserved. (3) The development of MDSCs was promoted and switched from an inactivated subset (CD33-CD11b+) to an activated subset (CD33+CD11b+). (4) The frequency of Foxp3+CD4+ regulatory T cells (Tregs) and CD24+CD38hi regulatory B cells was considerably increased in aGvHD patients, and Foxp3+CD8+ Tregs in cGvHD patients. (5) Proinflammatory cytokines like IL-1β, IL-6, IL-8, and TNF-α were significantly reduced. In summary, ECP constitutes an effective immunomodulatory therapy for patients with steroid-refractory/resistant GvHD without impairment of anti-viral/leukemia effects.
Collapse
Affiliation(s)
- Lei Wang
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Ming Ni
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany.,Department of Hematology, the Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | | | - Leopold Sellner
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Jean-Marc Hoffmann
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Brigitte Neuber
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Thomas Luft
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Ute Hegenbart
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Stefan Schönland
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Christian Kleist
- Department of Nuclear Medicine, University Clinic Heidelberg, Heidelberg, Germany
| | - Martin Sill
- Division Biostatistics, German Cancer Research Center, Heidelberg, Germany
| | - Bao-An Chen
- Department of Hematology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Patrick Wuchter
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany.,German Red Cross Blood Service, Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology Mannheim, Mannheim, Germany
| | - Volker Eckstein
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - William Krüger
- Department of Internal Medicine C, Haematology, Oncology, Stem Cell Transplantation, Palliative Care, University Clinic Greifswald, Greifswald, Germany
| | - Inken Hilgendorf
- Department of Internal Medicine II, University Clinic Jena, Jena, Germany
| | - Ronit Yerushalmi
- Hematology Division, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Arnon Nagler
- Hematology Division, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Anthony D Ho
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Peter Dreger
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Michael Schmitt
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Anita Schmitt
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| |
Collapse
|
93
|
Poe JC, Jia W, Di Paolo JA, Reyes NJ, Kim JY, Su H, Sundy JS, Cardones AR, Perez VL, Chen BJ, Chao NJ, Cardona DM, Saban DR, Sarantopoulos S. SYK inhibitor entospletinib prevents ocular and skin GVHD in mice. JCI Insight 2018; 3:122430. [PMID: 30282825 PMCID: PMC6237454 DOI: 10.1172/jci.insight.122430] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/29/2018] [Indexed: 12/15/2022] Open
Abstract
Graft-versus-host disease (GVHD) is a major complication of hematopoietic stem cell transplantation (HCT). The tyrosine kinase SYK contributes to both acute and chronic GVHD development, making it an attractive target for GVHD prevention. Entospletinib (ENTO) is a second-generation highly selective SYK inhibitor with a high safety profile. Potential utility of ENTO as GVHD prophylaxis in patients was examined using a preclinical mouse model of eye and skin GVHD and ENTO-compounded chow. We found that early SYK inhibition improved blood immune cell reconstitution in GVHD mice and prolonged survival, with 60% of mice surviving to day +120 compared with 10% of mice treated with placebo. Compared with mice receiving placebo, mice receiving ENTO had dramatic improvements in clinical eye scores, alopecia scores, and skin scores. Infiltrating SYK+ cells expressing B220 or F4/80, resembling SYK+ cells found in lichenoid skin lesions of chronic GVHD patients, were abundant in the skin of placebo mice but were rare in ENTO-treated mice. Thus, ENTO given early after HCT safely prevented GVHD.
Collapse
Affiliation(s)
- Jonathan C Poe
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, North Carolina, USA
| | - Wei Jia
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, North Carolina, USA
| | - Julie A Di Paolo
- Department of Biology, Gilead Sciences, Foster City, California, USA
| | - Nancy J Reyes
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina, USA
| | - Ji Yun Kim
- Department of Biology, Gilead Sciences, Foster City, California, USA
| | - Hsuan Su
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, North Carolina, USA
| | - John S Sundy
- Inflammation/Respiratory Section, Gilead Sciences, Foster City, California, USA
| | | | - Victor L Perez
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina, USA
| | - Benny J Chen
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, North Carolina, USA
| | - Nelson J Chao
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, North Carolina, USA
| | - Diana M Cardona
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Daniel R Saban
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina, USA
| | - Stefanie Sarantopoulos
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
94
|
New Answers to Old Conundrums: What Antibodies, Exosomes and Inflammasomes Bring to the Conversation. Canadian National Transplant Research Program International Summit Report. Transplantation 2018; 102:209-214. [PMID: 28731910 PMCID: PMC5802265 DOI: 10.1097/tp.0000000000001872] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Antibody-mediated injury is a major cause of allograft dysfunction and loss. Antibodies to ABH(O) blood group antigens are classic mediators of ABO-incompatible graft rejection, whereas donor-specific anti-HLA antibodies and, more recently, autoantibodies are appreciated as important contributors to allograft inflammation and dysfunction. In August 2016, the International Summit of the Canadian National Transplant Research Program focused on recent advances in the field of antibody-mediated rejection. Here, we describe work presented and discussed at the meeting, with a focus on 3 major themes: the importance of (1) natural antibodies and autoantibodies, (2) tissue injury-derived exosomes and autoimmunity, (3) inflammasome activation and innate immune responses in regulating allograft inflammation and dysfunction. Finally, we explore novel areas of therapeutic intervention that have recently emerged from these 3 major and overlapping fields of transplantation research.
Collapse
|
95
|
Fleischer T, Chang TT, Chiang JH, Yen HR. A Controlled Trial of Sheng-Yu-Tang for Post-Hematopoietic Stem Cell Transplantation Leukemia Patients: A Proposed Protocol and Insights From a Preliminary Pilot Study. Integr Cancer Ther 2018; 17:665-673. [PMID: 29431027 PMCID: PMC6142101 DOI: 10.1177/1534735418756736] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 01/09/2018] [Accepted: 01/09/2018] [Indexed: 11/15/2022] Open
Abstract
INTRODUCTION Hematopoietic stem cell transplantation has become a well-established treatment for hematologic disorders including acute leukemia. However, long-term survival rates following this procedure are still extremely low, due to posttransplantation relapse, infections, and graft-versus-host disease. We propose that adjunctive Chinese herbal medicine may benefit posttransplantation patients. In preparation for a randomized clinical trial, we conducted a pilot trial. Methods and Analysis: Between September 2015 and June 2017, 18 patients were consecutively enrolled at China Medical University Hospital and followed for up to 1 year. Fresh blood samples were obtained on a monthly basis, and immune reconstitution was analyzed. In addition to the standard-care treatment administered by their oncologist, a number of patients also received a Chinese herbal formula (Sheng-Yu-Tang) for up to 6 months. Results were used to improve on study protocol and estimate required sample size for a future randomized trial. Ethics and Dissemination: Study protocol was approved by the institutional review board of China Medical University Hospital (DMR-105-005), and all participants provided informed consent.
Collapse
Affiliation(s)
| | - Tung-Ti Chang
- China Medical University, Taichung,
Taiwan
- China Medical University Hospital,
Taichung, Taiwan
| | - Jen-Huai Chiang
- China Medical University, Taichung,
Taiwan
- China Medical University Hospital,
Taichung, Taiwan
| | - Hung-Rong Yen
- China Medical University, Taichung,
Taiwan
- China Medical University Hospital,
Taichung, Taiwan
- Asia University, Taichung, Taiwan
| |
Collapse
|
96
|
Stern L, McGuire H, Avdic S, Rizzetto S, Fazekas de St Groth B, Luciani F, Slobedman B, Blyth E. Mass Cytometry for the Assessment of Immune Reconstitution After Hematopoietic Stem Cell Transplantation. Front Immunol 2018; 9:1672. [PMID: 30093901 PMCID: PMC6070614 DOI: 10.3389/fimmu.2018.01672] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 07/05/2018] [Indexed: 12/31/2022] Open
Abstract
Mass cytometry, or Cytometry by Time-Of-Flight, is a powerful new platform for high-dimensional single-cell analysis of the immune system. It enables the simultaneous measurement of over 40 markers on individual cells through the use of monoclonal antibodies conjugated to rare-earth heavy-metal isotopes. In contrast to the fluorochromes used in conventional flow cytometry, metal isotopes display minimal signal overlap when resolved by single-cell mass spectrometry. This review focuses on the potential of mass cytometry as a novel technology for studying immune reconstitution in allogeneic hematopoietic stem cell transplant (HSCT) recipients. Reconstitution of a healthy donor-derived immune system after HSCT involves the coordinated regeneration of innate and adaptive immune cell subsets in the recipient. Mass cytometry presents an opportunity to investigate immune reconstitution post-HSCT from a systems-level perspective, by allowing the phenotypic and functional features of multiple cell populations to be assessed simultaneously. This review explores the current knowledge of immune reconstitution in HSCT recipients and highlights recent mass cytometry studies contributing to the field.
Collapse
Affiliation(s)
- Lauren Stern
- University of Sydney, Sydney, NSW, Australia.,Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,Discipline of Infectious Diseases and Immunology, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Helen McGuire
- University of Sydney, Sydney, NSW, Australia.,Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,Ramaciotti Facility for Human Systems Biology, University of Sydney, Sydney, NSW, Australia.,Discipline of Pathology, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
| | - Selmir Avdic
- University of Sydney, Sydney, NSW, Australia.,Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,Discipline of Infectious Diseases and Immunology, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | | | - Barbara Fazekas de St Groth
- University of Sydney, Sydney, NSW, Australia.,Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,Ramaciotti Facility for Human Systems Biology, University of Sydney, Sydney, NSW, Australia.,Discipline of Pathology, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
| | - Fabio Luciani
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Barry Slobedman
- University of Sydney, Sydney, NSW, Australia.,Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,Discipline of Infectious Diseases and Immunology, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Emily Blyth
- University of Sydney, Sydney, NSW, Australia.,Westmead Institute for Medical Research, University of Sydney, Sydney, NSW, Australia.,Blood and Marrow Transplant Unit, Westmead Hospital, Sydney, NSW, Australia.,Sydney Cellular Therapies Laboratory, Westmead, Sydney, NSW, Australia
| |
Collapse
|
97
|
Gooptu M, Kim HT, Chen YB, Rybka W, Artz A, Boyer M, Johnston L, McGuirk J, Shea TC, Jagasia M, Shaughnessy PJ, Reynolds CG, Fields M, Alyea EP, Ho VT, Glavin F, Dipersio JF, Westervelt P, Ritz J, Soiffer RJ. Effect of Antihuman T Lymphocyte Globulin on Immune Recovery after Myeloablative Allogeneic Stem Cell Transplantation with Matched Unrelated Donors: Analysis of Immune Reconstitution in a Double-Blind Randomized Controlled Trial. Biol Blood Marrow Transplant 2018; 24:2216-2223. [PMID: 30006305 DOI: 10.1016/j.bbmt.2018.07.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 07/03/2018] [Indexed: 02/06/2023]
Abstract
We recently conducted a randomized double-blind study in which we demonstrated that moderate/severe chronic graft-versus-host disease (cGVHD) but not cGVHD-free survival was reduced in patients receiving anti-T lymphocyte globulin (ATLG) versus placebo. In a companion study we performed immunophenotypic analysis to determine the impact of ATLG on immune reconstitution (IR) and to correlate IR with clinical outcomes. The randomized study (n = 254) included patients (aged 18 to 65 years) who underwent myeloablative transplants for acute myeloid leukemia, myelodysplastic syndrome, or acute lymphoblastic leukemia from HLA-matched unrelated donors. Ninety-one patients consented for the companion IR study (ATLG = 44, placebo = 47). Blood samples were collected on days 30, 100, 180, and 360 after hematopoietic cell transplantation (HCT), and multiparameter flow cytometry was performed in a blinded fashion. Reconstitution of CD3+ and CD4+ T cells was delayed up to 6 months post-HCT in the ATLG arm, whereas absolute regulatory T cell (Treg) (CD4+25+127-) numbers were lower only in the first 100 days. Analysis of the CD4+ Treg and conventional T cells (Tconv) (CD4+25-127+) compartments showed a profound absence of naive Tregs and Tconv in the first 100 days post-HCT, with very slow recovery for 1 year. B cell and natural killer cell recovery were similar in each arm. Higher absolute counts of CD3+, CD4+, CD8+ T, Tregs, and Tconv were associated with improved overall survival, progression-free survival, and nonrelapse mortality but not moderate/severe cGVHD. Although ATLG delays CD3+ and CD4+ T cell recovery post-transplant, it has a relative Treg sparing effect after the early post-HCT period, with possible implications for protection from cGVHD. ATLG severely compromises the generation of naive CD4+ cells (Treg and Tconv), potentially affecting the diversity of the TCR repertoire and T cell responses against malignancy and infection.
Collapse
Affiliation(s)
- Mahasweta Gooptu
- Dana-Farber Cancer Institute, Department of Hematologic Malignancies, Boston, Massachusetts USA
| | - Haesook T Kim
- Dana-Farber Cancer Institute, Department of Biostatistics and Computation Biology, Boston, Massachusetts USA
| | - Yi-Bin Chen
- Massachussetts General Hospital Department of Hematology/Oncology, Boston, Massachussetts, USA
| | - Witold Rybka
- Milton Hershey Medical Center, Department of Hematology/Oncology, Hershey, Pennsylvania, USA
| | - Andrew Artz
- University of Chicago, Comprehensive Cancer Center, Chicago, Illinois, USA. University of Utah, Pediatric Hematology/Oncology
| | - Michael Boyer
- Primary Children's Hospital, Salt Lake City, UT, USA
| | | | - Joseph McGuirk
- University of Kansas Medical Center, Department of Hematology/Oncology, Kansas City, Missouri, USA
| | - Thomas C Shea
- University of North Carolina, Chapel Hill, Division of Hematology/Oncology, North Carolina, USA
| | - Madan Jagasia
- Vanderbilt University Medical Center, Department of Hematology/Oncology, Nashville, TN, USA
| | | | - Carol G Reynolds
- Dana-Farber Cancer Institute, Department of Biostatistics and Computation Biology, Boston, Massachusetts USA
| | - Marie Fields
- Dana-Farber Cancer Institute, Department of Biostatistics and Computation Biology, Boston, Massachusetts USA
| | - Edwin P Alyea
- Dana-Farber Cancer Institute, Department of Biostatistics and Computation Biology, Boston, Massachusetts USA
| | - Vincent T Ho
- Dana-Farber Cancer Institute, Department of Biostatistics and Computation Biology, Boston, Massachusetts USA
| | | | - John F Dipersio
- BMT and Leukemia Program, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Peter Westervelt
- BMT and Leukemia Program, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jerome Ritz
- Dana-Farber Cancer Institute, Department of Biostatistics and Computation Biology, Boston, Massachusetts USA
| | - Robert J Soiffer
- Dana-Farber Cancer Institute, Department of Biostatistics and Computation Biology, Boston, Massachusetts USA
| |
Collapse
|
98
|
Wolff D, Greinix H, Lee SJ, Gooley T, Paczesny S, Pavletic S, Hakim F, Malard F, Jagasia M, Lawitschka A, Hansen JA, Pulanic D, Holler E, Dickinson A, Weissinger E, Edinger M, Sarantopoulos S, Schultz KR. Biomarkers in chronic graft-versus-host disease: quo vadis? Bone Marrow Transplant 2018; 53:832-837. [PMID: 29367715 PMCID: PMC6041126 DOI: 10.1038/s41409-018-0092-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 12/22/2017] [Indexed: 12/11/2022]
Abstract
Biomarkers are increasingly used for diagnosis and treatment of transplant-related complications including the first biomarker-driven interventional trials of acute graft-versus-host disease (GvHD). In contrast, the development of biomarkers of chronic GvHD (cGvHD) has lagged behind due to a broader variety of manifestations, overlap with acute GvHD, a greater variation in time to onset and maximum severity, and lack of sufficient patient numbers within prospective trials. An international workshop organized by a North-American and European consortium was held in Marseille in March 2017 with the goal to discuss strategies for future biomarker development to guide cGvHD therapy. As a result of this meeting, two areas were prioritized: the development of prognostic biomarkers for subsequent onset of moderate/severe cGvHD, and in parallel, the development of qualified clinical-grade assays for biomarker quantification. The most promising prognostic serum biomarkers are CXCL9, ST2, matrix metalloproteinase-3, osteopontin, CXCL10, CXCL11, and CD163. Urine-proteomics and cellular subsets (CD4+ T-cell subsets, NK cell subsets, and CD19+CD21low B cells) represent additional potential prognostic biomarkers of cGvHD. A joint effort is required to verify the results of numerous exploratory trials before any of the potential candidates is ready for validation and subsequent clinical application.
Collapse
Affiliation(s)
- D Wolff
- Department of Internal Medicine III, University Hospital of Regensburg, Regensburg, Germany.
| | - H Greinix
- Division of Haematology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - S J Lee
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - T Gooley
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - S Paczesny
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - S Pavletic
- Experimental Transplantation and Immunology Branch, Center of Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - F Hakim
- Experimental Transplantation and Immunology Branch, Center of Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - F Malard
- Hematology Department, Hôpital Saint-Antoine; Université Pierre & Marie Curie; and INSERM, Centre de Recherche Saint-Antoine, UMRS U938, Paris, France
| | - M Jagasia
- Department of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - A Lawitschka
- St. Anna Children's Hospital, Medical University Vienna, Vienna, Austria
| | - J A Hansen
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - D Pulanic
- Division of Hematology, Department of Internal Medicine, University Hospital Center Zagreb, and Medical School University of Zagreb, Zagreb, Croatia
- Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, Osijek, Croatia
| | - E Holler
- Department of Internal Medicine III, University Hospital of Regensburg, Regensburg, Germany
| | - A Dickinson
- Institute of Cellular Medicine, Newcastle University, Newcastle, UK
| | - E Weissinger
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - M Edinger
- Department of Internal Medicine III, University Hospital of Regensburg, Regensburg, Germany
| | - S Sarantopoulos
- Department of Medicine, Division of Hematological Malignancies & Cellular Therapy, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - K R Schultz
- Michael Cuccione Childhood Cancer Research Program, British Columbia Children's Hospital/University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
99
|
Bim regulates the survival and suppressive capability of CD8 + FOXP3 + regulatory T cells during murine GVHD. Blood 2018; 132:435-447. [PMID: 29769260 DOI: 10.1182/blood-2017-09-807156] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 05/09/2018] [Indexed: 12/29/2022] Open
Abstract
CD8+ Foxp3+ T cells (Tregs) are a potent regulatory population whose functional and ontological similarities to CD4+ Fox3+ T cells have not been well delineated. Using an experimental model of graft-versus-host disease (GVHD), we observed that CD8+ Tregs were significantly less potent than CD4+ Tregs for the suppression of GVHD. To define the mechanistic basis for this observation, we examined the T-cell repertoire and the transcriptional profile of in vivo-derived CD4+ and CD8+ Tregs that emerged early during this disease. Polyclonal and alloantigen-induced CD8+ Tregs had repertoire diversity that was similar to that of conventional CD8+ T cells, indicating that a restricted repertoire was not the proximate cause of decreased suppression. Transcriptional profiling revealed that CD8+ Tregs possessed a canonical Treg transcriptional signature that was similar to that observed in CD4+ Tregs, yet distinct from conventional CD8+ T cells. Pathway analysis, however, demonstrated that CD8+ Tregs had differential gene expression in pathways involved in cell death and survival. This was further confirmed by detailed mRNA sequence analysis and protein expression studies, which demonstrated that CD8+ Tregs had increased expression of Bim and reduced expression of Mcl-1. Transplantation with CD8+ Foxp3+ Bim-/- Tregs resulted in prolonged Treg survival and reduced GVHD lethality compared with wild-type CD8+ Tregs, providing functional confirmation that increased expression of Bim was responsible for reduced in vivo efficacy. Thus, Bim regulates the survival and suppressive capability of CD8+ Tregs, which may have implications for their use in regulatory T-cell therapy.
Collapse
|
100
|
Ren HG, Adom D, Paczesny S. The search for drug-targetable diagnostic, prognostic and predictive biomarkers in chronic graft-versus-host disease. Expert Rev Clin Immunol 2018; 14:389-404. [PMID: 29629613 DOI: 10.1080/1744666x.2018.1463159] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Chronic graft-versus-host disease (cGVHD) continues to be the leading cause of late morbidity and mortality after allogeneic hematopoietic stem cell transplantation (allo-HSCT), which is an increasingly applied curative method for both benign and malignant hematologic disorders. Biomarker identification is crucial for the development of noninvasive and cost-effective cGVHD diagnostic, prognostic, and predictive test for use in clinic. Furthermore, biomarkers may help to gain a better insight on ongoing pathophysiological processes. The recent widespread application of omics technologies including genomics, transcriptomics, proteomics and cytomics provided opportunities to discover novel biomarkers. Areas covered: This review focuses on biomarkers identified through omics that play a critical role in target identification for drug development, and that were verified in at least two independent cohorts. It also summarizes the current status on omics tools used to identify these useful cGVHD targets. We briefly list the biomarkers identified and verified so far. We further address challenges associated to their exploitation and application in the management of cGVHD patients. Finally, insights on biomarkers that are drug targetable and represent potential therapeutic targets are discussed. Expert commentary: We focus on biomarkers that play an essential role in target identification.
Collapse
Affiliation(s)
- Hong-Gang Ren
- a Department of Pediatrics , Indiana University , Indianapolis , IN , USA.,b Department of Microbiology Immunology , Indiana University , Indianapolis , IN , USA.,c Melvin and Bren Simon Cancer Center , Indiana University , Indianapolis , IN , USA
| | - Djamilatou Adom
- a Department of Pediatrics , Indiana University , Indianapolis , IN , USA.,b Department of Microbiology Immunology , Indiana University , Indianapolis , IN , USA.,c Melvin and Bren Simon Cancer Center , Indiana University , Indianapolis , IN , USA
| | - Sophie Paczesny
- a Department of Pediatrics , Indiana University , Indianapolis , IN , USA.,b Department of Microbiology Immunology , Indiana University , Indianapolis , IN , USA.,c Melvin and Bren Simon Cancer Center , Indiana University , Indianapolis , IN , USA
| |
Collapse
|