1
|
Fu H, Yu J. [Pathogenesis and treatment progression of myelodysplastic syndrome combined with Behçet 's syndrome]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2025; 54:131-139. [PMID: 39949137 PMCID: PMC11956866 DOI: 10.3724/zdxbyxb-2024-0150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 01/05/2025] [Indexed: 02/25/2025]
Abstract
Myelodysplastic syndromes (MDS) are clonal hematopoietic neoplasms characterized by chronic cytopenias and abnormal cell morphology, with a propensity of progressing to bone marrow failure or acute myeloid leukemia. Behçet's syndrome is a systemic vasculitis characterized by recurrent oral ulcers, skin lesions, and ocular inflammation. In recent years, an increasing number of clinical cases with coexistence of MDS and Behçet's syndrome have been reported, suggesting a potential pathological relationship between these conditions. Abnormal immune cell activation, dysregulated cytokine secretion, and cytogenetic alterations are thought to play critical roles in the pathogenesis of MDS combined with Behçet's syndrome. Currently, treatment strategies for MDS combined with Behçet's syndrome are primarily individualized and include immunosuppressive therapy, cytotoxic drug therapy, targeted therapy, and hematopoietic stem cell transplantation. However, due to the limited number of case reports and insufficient research on the underlying mechanisms, selecting appropriate treatment options remains challenging. This article reviews the pathogenesis and interrelationships of MDS combined with Behçet's syndrome and summarizes recent advancements in treatment strategies, providing a reference for clinical management and further researches on related mechanisms.
Collapse
Affiliation(s)
- Hui Fu
- Department of Hematology and Oncology, Taizhou Hospital of Zhejiang Province, Taizhou 317000, Zhejiang Province, China.
| | - Jian Yu
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
2
|
Lei R, Liu X, Wu Y, Song J, Lv X, Liu J, Guo D, Li H, Fan S. Application of allogeneic hematopoietic stem cell transplantation to treat Behcet's disease with myelodysplastic syndrome: a case report and literature review. Ann Hematol 2024:10.1007/s00277-024-06122-y. [PMID: 39611877 DOI: 10.1007/s00277-024-06122-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024]
Abstract
Trisomy 8 is a frequent chromosomal abnormality that occurs among patients with myelodysplastic syndrome (MDS). This abnormality is more common among MDS patients with concurrent Behcet's disease (BD) than among patients with MDS alone. This comorbidity is also associated with a higher prevalence of intestinal ulcers. In this case report, we describe a 45-year-old woman who was diagnosed with BD at age 35. Eight years later, her Behcet's disease relapsed with trisomy 8-positive MDS. After the disease was controlled by azacitidine, the patient received allogeneic hematopoietic stem cell transplantation (allo-HSCT), which involved the use of both peripheral blood stem cells and bone marrow stem cells. Although the patient experienced grave oral mucositis and chronic intestinal graft-versus-host disease (GVHD), remission of both MDS and BD was maintained. This case highlights the potential efficacy of allo-HSCT in treating patients with refractory autoimmune diseases complicated by MDS, particularly patients with trisomy 8 abnormalities. We also discuss the immunological implications and suggest allo-HSCT as a viable curative option for patients with BD and MDS.
Collapse
Affiliation(s)
- RuiQi Lei
- Division of Hematology, Department of Medicine, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nan Gang District, Harbin, 150001, Heilongjiang Province, China
| | - Xin Liu
- Division of Hematology, Department of Medicine, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nan Gang District, Harbin, 150001, Heilongjiang Province, China
| | - YiTing Wu
- Division of Hematology, Department of Medicine, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nan Gang District, Harbin, 150001, Heilongjiang Province, China
| | - JingYu Song
- Division of Hematology, Department of Medicine, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nan Gang District, Harbin, 150001, Heilongjiang Province, China
| | - XiaoLi Lv
- Division of Hematology, Department of Medicine, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nan Gang District, Harbin, 150001, Heilongjiang Province, China
| | - Jie Liu
- Division of Hematology, Department of Medicine, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nan Gang District, Harbin, 150001, Heilongjiang Province, China
| | - Dan Guo
- Division of Hematology, Department of Medicine, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nan Gang District, Harbin, 150001, Heilongjiang Province, China
| | - HuiBo Li
- Division of Hematology, Department of Medicine, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nan Gang District, Harbin, 150001, Heilongjiang Province, China.
| | - ShengJin Fan
- Division of Hematology, Department of Medicine, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nan Gang District, Harbin, 150001, Heilongjiang Province, China.
- NHC Key Laboratory of Cell Transplantation, the First Affiliated Hospital, Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
3
|
Tazoe K, Harada N, Makuuchi Y, Kuno M, Takakuwa T, Okamura H, Hirose A, Nakamae M, Nishimoto M, Nakashima Y, Koh H, Hino M, Nakamae H. Systemic inflammatory autoimmune disease before allogeneic hematopoietic stem cell transplantation is a risk factor for death in patients with myelodysplastic syndrome or chronic myelomonocytic leukemia. Ann Hematol 2024; 103:2059-2072. [PMID: 38662207 DOI: 10.1007/s00277-024-05772-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 04/20/2024] [Indexed: 04/26/2024]
Abstract
Myelodysplastic syndrome (MDS) is well known to be complicated by systemic inflammatory autoimmune disease (SIADs). However, it remains unclear how the prognosis after allogenic hematopoietic stem cell transplantation (allo-HSCT) in patients with MDS is impacted by SIADs that occur before allo-HSCT. Therefore, we hypothesized that SIADs before allo-HSCT may be a risk factor for negative outcomes after allo-HSCT in patients with MDS. We conducted a single-center, retrospective, observational study of sixty-nine patients with MDS or chronic myelomonocytic leukemia who underwent their first allo-HCT. Fourteen of the patients had SIADs before allo-HSCT. In multivariate analysis, the presence of SIADs before allo-HSCT was an independent risk factor for overall survival (HR, 3.36, 95% confidence interval: 1.34-8.42, p = 0.009). Endothelial dysfunction syndrome was identified in five of 14 patients with SIADs who required immunosuppressive therapy or intensive chemotherapy, and notably, all patients with uncontrollable SIADs at allo-HSCT developed serious endothelial dysfunction syndrome and died in the early phase after allo-HSCT. The development of SIADs in the context of MDS is thought to reflect the degree of dysfunction of hematopoietic cells in MDS and suggests a higher risk of disease progression. In addition, MDS patients with SIADs before allo-HSCT are considered to be at higher risk of endothelial dysfunction syndrome because of preexisting vascular endothelial dysfunction due to SIADs. In conclusion, SIADs before allo-HSCT constitute an independent risk factor for death in MDS patients undergoing allo-HSCT.
Collapse
Affiliation(s)
- Kumiyo Tazoe
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
- Department of Hematology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Naonori Harada
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan.
- Department of Hematology, Fuchu Hospital, Osaka, Japan.
| | - Yosuke Makuuchi
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Masatomo Kuno
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Teruhito Takakuwa
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Hiroshi Okamura
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Asao Hirose
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Mika Nakamae
- Department of Clinical Laboratory, Osaka Metropolitan University Hospital, Osaka, Japan
- Department of Laboratory Medicine and Medical Informatics, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Mitsutaka Nishimoto
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Yasuhiro Nakashima
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Hideo Koh
- Department of Preventive Medicine and Environmental Health, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Masayuki Hino
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Hirohisa Nakamae
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
4
|
Rodriguez-Sevilla JJ, Colla S. T-cell dysfunctions in myelodysplastic syndromes. Blood 2024; 143:1329-1343. [PMID: 38237139 DOI: 10.1182/blood.2023023166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/22/2023] [Accepted: 01/12/2024] [Indexed: 03/25/2024] Open
Abstract
ABSTRACT Escape from immune surveillance is a hallmark of cancer. Immune deregulation caused by intrinsic and extrinsic cellular factors, such as altered T-cell functions, leads to immune exhaustion, loss of immune surveillance, and clonal proliferation of tumoral cells. The T-cell immune system contributes to the pathogenesis, maintenance, and progression of myelodysplastic syndrome (MDS). Here, we comprehensively reviewed our current biological knowledge of the T-cell compartment in MDS and recent advances in the development of immunotherapeutic strategies, such as immune checkpoint inhibitors and T-cell- and antibody-based adoptive therapies that hold promise to improve the outcome of patients with MDS.
Collapse
Affiliation(s)
| | - Simona Colla
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
5
|
Leone S, Rubino V, Palatucci AT, Giovazzino A, Carriero F, Cerciello G, Pane F, Ruggiero G, Terrazzano G. Bone Marrow CD3 + CD56 + Regulatory T lymphocytes (T R3-56 cells) are inversely associated with activation and expansion of Bone Marrow cytotoxic T cells in IPSS-R very-low/low risk MDS patients. Eur J Haematol 2022; 109:398-405. [PMID: 35775392 PMCID: PMC9543123 DOI: 10.1111/ejh.13822] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 12/04/2022]
Abstract
Background Emergence of dysplastic haematopoietic precursor/s, cytopenia and variable leukaemia risk characterise myelodysplastic syndromes (MDS). Impaired immune‐regulation, preferentially affecting cytotoxic T cells (CTL), has been largely observed in MDS. Recently, we described the TR3−56 T cell subset, characterised by the co‐expression of CD3 and CD56, as a novel immune‐regulatory population, able to modulate cytotoxic functions. Here, we address the involvement of TR3−56 cells in MDS pathogenesis/progression. Objectives To analyse the relationship between TR3−56 and CTL activation/expansion in bone marrow (BM) of very‐low/low‐risk MDS subjects. Methods Peripheral blood and BM specimens, obtained at disease onset in a cohort of 58 subjects, were analysed by immune‐fluorescence and flow cytometry, to preserve the complexity of the biological sample. Results We observed that a trend‐increase of BM TR3−56 in high/very‐high MDS stage, as compared with very‐low/low group, associates with a decreased activation of BM resident CTL; significant correlation of TR3−56 with BM blasts has been also revealed. In addition, in very‐low/low‐risk subjects the TR3−56 amount in BM inversely correlates with the presence of activated BM CTL showing a skewed Vβ T‐cell repertoire. Conclusions These data add TR3−56 to the immune‐regulatory network involved in MDS pathogenesis/progression. Better knowledge of the immune‐mediated processes associated with the disease might improve MDS clinical management.
Collapse
Affiliation(s)
- Stefania Leone
- Divisione di Ematologia, Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli "Federico II"
| | - Valentina Rubino
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli "Federico II", Napoli, Italy
| | | | - Angela Giovazzino
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli "Federico II", Napoli, Italy
| | - Flavia Carriero
- Ph.D course in Science, Università della Basilicata, Via dell'Ateneo Lucano, Potenza, Italy
| | - Giuseppe Cerciello
- Divisione di Ematologia, Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli "Federico II"
| | - Fabrizio Pane
- Divisione di Ematologia, Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli "Federico II"
| | - Giuseppina Ruggiero
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli "Federico II", Napoli, Italy
| | - Giuseppe Terrazzano
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli "Federico II", Napoli, Italy.,Dipartimento di Scienze, Università della Basilicata, Potenza, Italy
| |
Collapse
|
6
|
Tria FP, Ang DC, Fan G. Myelodysplastic Syndrome: Diagnosis and Screening. Diagnostics (Basel) 2022; 12:1581. [PMID: 35885487 PMCID: PMC9319204 DOI: 10.3390/diagnostics12071581] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 11/16/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are heterogeneous groups of clonal myeloid disorders characterized by unexplained persistent peripheral blood (PB) cytopenia(s) of one or more of the hematopoietic lineages, or bone marrow (BM) morphologic dysplasia in hematopoietic cells, recurrent genetic abnormalities, and an increased risk of progression to acute myeloid leukemia (AML). In the past several years, diagnostic, prognostic, and therapeutic approaches have substantially improved with the development of Next Generation Sequencing (NGS) diagnostic testing and new medications. However, there is no single diagnostic parameter specific for MDS, and correlations with clinical information, and laboratory test findings are needed to reach the diagnosis.
Collapse
Affiliation(s)
- Francisco P. Tria
- Section of Cellular Immunology and Molecular Pathology, Institute of Pathology, St. Luke’s Medical Center—Global City, Taguig 1634, Metro Manila, Philippines; (F.P.T.IV); (D.C.A.)
| | - Daphne C. Ang
- Section of Cellular Immunology and Molecular Pathology, Institute of Pathology, St. Luke’s Medical Center—Global City, Taguig 1634, Metro Manila, Philippines; (F.P.T.IV); (D.C.A.)
| | - Guang Fan
- Department of Hematopathology, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
7
|
Votavova H, Belickova M. Hypoplastic myelodysplastic syndrome and acquired aplastic anemia: Immune‑mediated bone marrow failure syndromes (Review). Int J Oncol 2021; 60:7. [PMID: 34958107 PMCID: PMC8727136 DOI: 10.3892/ijo.2021.5297] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/01/2021] [Indexed: 11/06/2022] Open
Abstract
Hypoplastic myelodysplastic syndrome (hMDS) and aplastic anemia (AA) are rare hematopoietic disorders characterized by pancytopenia with hypoplastic bone marrow (BM). hMDS and idiopathic AA share overlapping clinicopathological features, making a diagnosis very difficult. The differential diagnosis is mainly based on the presence of dysgranulopoiesis, dysmegakaryocytopoiesis, an increased percentage of blasts, and abnormal karyotype, all favouring the diagnosis of hMDS. An accurate diagnosis has important clinical implications, as the prognosis and treatment can be quite different for these diseases. Patients with hMDS have a greater risk of neoplastic progression, a shorter survival time and a lower response to immunosuppressive therapy compared with patients with AA. There is compelling evidence that these distinct clinical entities share a common pathophysiology based on the damage of hematopoietic stem and progenitor cells (HSPCs) by cytotoxic T cells. Expanded T cells overproduce proinflammatory cytokines (interferon-γ and tumor necrosis factor-α), resulting in decreased proliferation and increased apoptosis of HSPCs. The antigens that trigger this abnormal immune response are not known, but potential candidates have been suggested, including Wilms tumor protein 1 and human leukocyte antigen class I molecules. Our understanding of the molecular pathogenesis of these BM failure syndromes has been improved by next-generation sequencing, which has enabled the identification of a large spectrum of mutations. It has also brought new challenges, such as the interpretation of variants of uncertain significance and clonal hematopoiesis of indeterminate potential. The present review discusses the main clinicopathological differences between hMDS and acquired AA, focuses on the molecular background and highlights the importance of molecular testing.
Collapse
Affiliation(s)
- Hana Votavova
- Department of Genomics, Institute of Hematology and Blood Transfusion, Prague 128 00, Czech Republic
| | - Monika Belickova
- Department of Genomics, Institute of Hematology and Blood Transfusion, Prague 128 00, Czech Republic
| |
Collapse
|
8
|
Chee L, Ritchie D, Ludford-Menting M, Ripley J, Chung J, Park D, Norton S, Kenealy M, Koldej R. Dysregulation of immune cell and cytokine signaling correlates with clinical outcomes in myelodysplastic syndrome (MDS). Eur J Haematol 2021; 108:342-353. [PMID: 34963023 DOI: 10.1111/ejh.13742] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/19/2021] [Accepted: 12/21/2021] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Myelodysplastic syndromes (MDS) are characterized by ineffective hematopoiesis. Although hypomethylating agents (HMA) have improved survival in higher-risk MDS, most patients eventually succumb to progressive disease. Utilizing samples collected prospectively from three MDS clinical trials, we analyzed genetic and immunological biomarkers and correlated them with clinical outcomes. METHODS 154 samples were analyzed from 133 de novo MDS patients for T-cell and myeloid cell immunophenotyping and gene expression analysis. Treatments were with HMA or immunomodulatory drug (IMiD) alone or in combination. RESULTS We observed differences in immune cell subsets between lower and higher risk IPSS groups with NKT cells, MDSCs, intermediate-proinflammatory and non-classical monocytes being higher in the latter group while naïve CD4+ T-cells were reduced. Intermediate-proinflammatory monocytes were increased in non-responders and those failing to achieve at least a hematological improvement. Pro-inflammatory NKT cells were increased at diagnosis for patients failing to derive clinical benefit after 12 months of treatment. Gene expression analysis of paired bone marrow (BM) colony-forming units (CFUs) from diagnosis and 4 cycles post-treatment confirmed that genes involved in cytokine signaling were downregulated in C4 normal colonies. CONCLUSIONS These findings support the central roles of dysregulation in innate immunity and inflammatory signaling in the pathogenesis of MDS which correlated with clinical outcomes post-treatment.
Collapse
Affiliation(s)
- L Chee
- Australian Cancer Research Foundation (ACRF) Translational Laboratory, The Royal Melbourne Hospital, Melbourne, VIC, 3000, Australia.,Department of Clinical Haematology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Parkville, VIC, 3050, Australia.,Department of Medicine, The University of Melbourne, Parkville, VIC, 3050, Australia.,The Australasian Leukaemia and Lymphoma Group, Richmond, VIC, 3121, Australia
| | - D Ritchie
- Australian Cancer Research Foundation (ACRF) Translational Laboratory, The Royal Melbourne Hospital, Melbourne, VIC, 3000, Australia.,Department of Clinical Haematology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Parkville, VIC, 3050, Australia.,Department of Medicine, The University of Melbourne, Parkville, VIC, 3050, Australia.,The Australasian Leukaemia and Lymphoma Group, Richmond, VIC, 3121, Australia
| | - M Ludford-Menting
- Australian Cancer Research Foundation (ACRF) Translational Laboratory, The Royal Melbourne Hospital, Melbourne, VIC, 3000, Australia
| | - J Ripley
- Australian Cancer Research Foundation (ACRF) Translational Laboratory, The Royal Melbourne Hospital, Melbourne, VIC, 3000, Australia.,Liverpool Hospital, Liverpool, NSW, 2170, Australia
| | - J Chung
- Melbourne Bioinformatics, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - D Park
- Melbourne Bioinformatics, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - S Norton
- Nanix Limited, Dunedin, New Zealand
| | - M Kenealy
- The Australasian Leukaemia and Lymphoma Group, Richmond, VIC, 3121, Australia.,Cabrini Hospital, Malvern, VIC, 3144, Australia.,Monash University, Clayton, VIC, 3800, Australia
| | - R Koldej
- Australian Cancer Research Foundation (ACRF) Translational Laboratory, The Royal Melbourne Hospital, Melbourne, VIC, 3000, Australia.,Department of Medicine, The University of Melbourne, Parkville, VIC, 3050, Australia
| |
Collapse
|
9
|
Camacho V, Kuznetsova V, Welner RS. Inflammatory Cytokines Shape an Altered Immune Response During Myeloid Malignancies. Front Immunol 2021; 12:772408. [PMID: 34804065 PMCID: PMC8595317 DOI: 10.3389/fimmu.2021.772408] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/19/2021] [Indexed: 12/14/2022] Open
Abstract
The immune microenvironment is a critical driver and regulator of leukemic progression and hematological disease. Recent investigations have demonstrated that multiple immune components play a central role in regulating hematopoiesis, and dysfunction at the immune cell level significantly contributes to neoplastic disease. Immune cells are acutely sensitive to remodeling by leukemic inflammatory cytokine exposure. Importantly, immune cells are the principal cytokine producers in the hematopoietic system, representing an untapped frontier for clinical interventions. Due to a proinflammatory cytokine environment, dysregulation of immune cell states is a hallmark of hematological disease and neoplasia. Malignant immune adaptations have profound effects on leukemic blast proliferation, disease propagation, and drug-resistance. Conversely, targeting the immune landscape to restore hematopoietic function and limit leukemic expansion may have significant therapeutic value. Despite the fundamental role of the immune microenvironment during the initiation, progression, and treatment response of hematological disease, a detailed examination of how leukemic cytokines alter immune cells to permit, promote, or inhibit leukemia growth is lacking. Here we outline an immune-based model of leukemic transformation and highlight how the profound effect of immune alterations on the trajectory of malignancy. The focus of this review is to summarize current knowledge about the impacts of pro- and anti-inflammatory cytokines on immune cells subsets, their modes of action, and immunotherapeutic approaches with the potential to improve clinical outcomes for patients suffering from hematological myeloid malignancies.
Collapse
Affiliation(s)
- Virginia Camacho
- Department of Medicine, Division of Hematology/Oncology, O'Neal Comprehensive Cancer Center at the University of Alabama at Birmingham, Birmingham, AL, United States
| | - Valeriya Kuznetsova
- Department of Medicine, Division of Hematology/Oncology, O'Neal Comprehensive Cancer Center at the University of Alabama at Birmingham, Birmingham, AL, United States
| | - Robert S Welner
- Department of Medicine, Division of Hematology/Oncology, O'Neal Comprehensive Cancer Center at the University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
10
|
Buchrits S, Kozlovoski D, Gafter-Gvili A, Raanani P, Hayman L, Revyako I, Bernstine H, Wolach O, Shimony S. Autoimmune and Inflammatory Manifestations Associated with Acute Myeloid Leukemia with Trisomy 8 - Case Series and Review of the Literature. Eur J Haematol 2021; 108:199-203. [PMID: 34748235 DOI: 10.1111/ejh.13725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/02/2021] [Indexed: 11/27/2022]
Abstract
AML can be associated with autoimmune or inflammatory phenomena (AIP) occurring prior, concomitantly or after its diagnosis. Trisomy 8 is one of the most common cytogenetic abnormalities associated with AML. We describe three patients with AML, trisomy 8 and associated AIP and review the known literature on this association. All of our patients had major symptomatic relief when treated with leukemia-directed therapy and corticosteroids. AIP in AML may be an underdiagnosed phenomenon, particularly in patients with trisomy 8.
Collapse
Affiliation(s)
- Shira Buchrits
- Internal Medicine Department A, Rabin Medical Center, Beilinson Hospital, Petah-Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Dror Kozlovoski
- Internal Medicine Department A, Rabin Medical Center, Beilinson Hospital, Petah-Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Anat Gafter-Gvili
- Internal Medicine Department A, Rabin Medical Center, Beilinson Hospital, Petah-Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel.,Institute of Haematology, Davidoff Cancer Center, Rabin Medical Center, Beilinson Hospital, Petah-Tikva, Israel
| | - Pia Raanani
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel.,Institute of Haematology, Davidoff Cancer Center, Rabin Medical Center, Beilinson Hospital, Petah-Tikva, Israel
| | - Lucille Hayman
- Pathology Department, Rabin Medical Center, Beilinson Hospital, Petah Tikva, Israel
| | - Igor Revyako
- Pathology Department, Rabin Medical Center, Beilinson Hospital, Petah Tikva, Israel
| | - Hanna Bernstine
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel.,Nuclear Medicine Department, Rabin Medical Center, Beilinson Hospital, Petah Tikva, Israel
| | - Ofir Wolach
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel.,Institute of Haematology, Davidoff Cancer Center, Rabin Medical Center, Beilinson Hospital, Petah-Tikva, Israel
| | - Shai Shimony
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel.,Institute of Haematology, Davidoff Cancer Center, Rabin Medical Center, Beilinson Hospital, Petah-Tikva, Israel
| |
Collapse
|
11
|
Kapoor S, Champion G, Basu A, Mariampillai A, Olnes MJ. Immune Therapies for Myelodysplastic Syndromes and Acute Myeloid Leukemia. Cancers (Basel) 2021; 13:5026. [PMID: 34638510 PMCID: PMC8507987 DOI: 10.3390/cancers13195026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 11/16/2022] Open
Abstract
Myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) are hematologic malignancies arising from the bone marrow. Despite recent advances in treating these diseases, patients with higher-risk MDS and AML continue to have a poor prognosis with limited survival. It has long been recognized that there is an immune component to the pathogenesis of MDS and AML, but until recently, immune therapies have played a limited role in treating these diseases. Immune suppressive therapy exhibits durable clinical responses in selected patients with MDS, but the question of which patients are most suitable for this treatment remains unclear. Over the past decade, there has been remarkable progress in identifying genomic features of MDS and AML, which has led to an improved discernment of the molecular pathogenesis of these diseases. An improved understanding of immune and inflammatory molecular mechanisms of MDS and AML have also recently revealed novel therapeutic targets. Emerging treatments for MDS and AML include monoclonal antibodies such as immune checkpoint inhibitors, bispecific T-cell-engaging antibodies, antibody drug conjugates, vaccine therapies, and cellular therapeutics including chimeric antigen receptor T-cells and NK cells. In this review, we provide an overview of the current understanding of immune dysregulation in MDS and AML and an update on novel immune therapies for these bone marrow malignancies.
Collapse
Affiliation(s)
- Sargam Kapoor
- Hematology and Medical Oncology, Alaska Native Tribal Health Consortium, 3900 Ambassador Dr., Anchorage, AK 99508, USA; (S.K.); (A.B.); (A.M.)
- School of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA;
| | - Grace Champion
- School of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA;
| | - Aparna Basu
- Hematology and Medical Oncology, Alaska Native Tribal Health Consortium, 3900 Ambassador Dr., Anchorage, AK 99508, USA; (S.K.); (A.B.); (A.M.)
| | - Anu Mariampillai
- Hematology and Medical Oncology, Alaska Native Tribal Health Consortium, 3900 Ambassador Dr., Anchorage, AK 99508, USA; (S.K.); (A.B.); (A.M.)
- School of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA;
| | - Matthew J. Olnes
- Hematology and Medical Oncology, Alaska Native Tribal Health Consortium, 3900 Ambassador Dr., Anchorage, AK 99508, USA; (S.K.); (A.B.); (A.M.)
- School of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA;
- WWAMI School of Medical Education, University of Alaska Anchorage, 3211 Providence Drive, Anchorage, AK 99508, USA
| |
Collapse
|
12
|
Immunologic effects on the haematopoietic stem cell in marrow failure. Best Pract Res Clin Haematol 2021; 34:101276. [PMID: 34404528 DOI: 10.1016/j.beha.2021.101276] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 02/07/2023]
Abstract
Acquired bone marrow failure (BMF) syndromes comprise a diverse group of diseases with variable clinical manifestations but overlapping features of immune activation, resulting in haematopoietic stem and progenitor cells (HSPC) damage and destruction. This review focuses on clinical presentation, pathophysiology, and treatment of four BMF: acquired aplastic anaemia, large granular lymphocytic leukaemia, paroxysmal nocturnal haemoglobinuria, and hypoplastic myelodysplastic syndrome. Autoantigens are speculated to be the inciting event that result in immune activation in all of these diseases, but specific pathogenic antigens have not been identified. Oligoclonal cytotoxic T cell expansion and an active role of proinflammatory cytokines, primarily interferon gamma (IFN-γ) and tumor necrosis factor alpha (TNF-α), are two main contributors to HSPC growth inhibition and apoptosis in BMF. Emerging evidence also suggests involvement of the innate immune system.
Collapse
|
13
|
Asian Population Is More Prone to Develop High-Risk Myelodysplastic Syndrome, Concordantly with Their Propensity to Exhibit High-Risk Cytogenetic Aberrations. Cancers (Basel) 2021; 13:cancers13030481. [PMID: 33513838 PMCID: PMC7865620 DOI: 10.3390/cancers13030481] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/13/2021] [Accepted: 01/19/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary The world population is genetically and environmentally diverse. In particular, genetic differences related to an ethnic factor may underlie differences in cancer phenotypic expression. Therefore, we compared the epidemiology, and the clinical, biological and genetic characteristics of myelodysplastic syndrome (MDS) between Asian and Western countries. Our results show substantial differences in the incidence and age of onset between Asian and Western MDS patients. A higher proportion of Asian MDS patients fall into the high- and very-high risk prognostic MDS groups. This finding is supported by the identification of a higher proportion of high-risk cytogenetic aberrations in Asian MDS patients. However, the survival rate is similar for Western and Asian MDS patients. Our findings may impact the clinical management as well as the strategy of clinical trials targeting those genetic aberrations and mutations depending on the world area where they are run. Abstract This study explores the hypothesis that genetic differences related to an ethnic factor may underlie differences in phenotypic expression of myelodysplastic syndrome (MDS). First, to identify clear ethnic differences, we systematically compared the epidemiology, and the clinical, biological and genetic characteristics of MDS between Asian and Western countries over the last 20 years. Asian MDS cases show a 2- to 4-fold lower incidence and a 10-year younger age of onset compared to the Western cases. A higher proportion of Western MDS patients fall into the very low- and low-risk categories while the intermediate, high and very high-risk groups are more represented in Asian MDS patients according to the Revised International Prognostic Scoring System. Next, we investigated whether differences in prognostic risk scores could find their origin in differential cytogenetic profiles. We found that 5q deletion (del(5q)) aberrations and mutations in TET2, SF3B1, SRSF2 and IDH1/2 are more frequently reported in Western MDS patients while trisomy 8, del(20q), U2AF1 and ETV6 mutations are more frequent in Asian MDS patients. Treatment approaches differ between Western and Asian countries owing to the above discrepancies, but the overall survival rate within each prognostic group is similar for Western and Asian MDS patients. Altogether, our study highlights greater risk MDS in Asians supported by their cytogenetic profile.
Collapse
|
14
|
Giudice V, Cardamone C, Triggiani M, Selleri C. Bone Marrow Failure Syndromes, Overlapping Diseases with a Common Cytokine Signature. Int J Mol Sci 2021; 22:ijms22020705. [PMID: 33445786 PMCID: PMC7828244 DOI: 10.3390/ijms22020705] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/06/2021] [Accepted: 01/09/2021] [Indexed: 12/19/2022] Open
Abstract
Bone marrow failure (BMF) syndromes are a heterogenous group of non-malignant hematologic diseases characterized by single- or multi-lineage cytopenia(s) with either inherited or acquired pathogenesis. Aberrant T or B cells or innate immune responses are variously involved in the pathophysiology of BMF, and hematological improvement after standard immunosuppressive or anti-complement therapies is the main indirect evidence of the central role of the immune system in BMF development. As part of this immune derangement, pro-inflammatory cytokines play an important role in shaping the immune responses and in sustaining inflammation during marrow failure. In this review, we summarize current knowledge of cytokine signatures in BMF syndromes.
Collapse
Affiliation(s)
- Valentina Giudice
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (V.G.); (C.C.); (C.S.)
- Clinical Pharmacology, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, 84131 Salerno, Italy
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, 84131 Salerno, Italy
| | - Chiara Cardamone
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (V.G.); (C.C.); (C.S.)
- Internal Medicine and Clinical Immunology, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, 84131 Salerno, Italy
| | - Massimo Triggiani
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (V.G.); (C.C.); (C.S.)
- Internal Medicine and Clinical Immunology, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, 84131 Salerno, Italy
- Correspondence: ; Tel.: +39-089-672810
| | - Carmine Selleri
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (V.G.); (C.C.); (C.S.)
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, 84131 Salerno, Italy
| |
Collapse
|
15
|
Hypoplastic Myelodysplastic Syndromes: Just an Overlap Syndrome? Cancers (Basel) 2021; 13:cancers13010132. [PMID: 33401595 PMCID: PMC7795441 DOI: 10.3390/cancers13010132] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/29/2020] [Accepted: 12/29/2020] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Hypoplastic myelodysplastic syndromes (hMDS) represent a diagnostic conundrum. They share morphologic and clinical features of both MDS (dysplasia, genetic lesions and cytopenias) and aplastic anemia (AA; i.e., hypocellularity and autoimmunity) and are not comprised in the last WHO classification. In this review we recapitulate the main clinical, pathogenic and therapeutic aspects of hypo-MDS and discuss why they deserve to be distinguished from normo/hypercellular MDS and AA. We conclude that hMDS may present in two phenotypes: one more proinflammatory and autoimmune, more similar to AA, responding to immunosuppression; and one MDS-like dominated by genetic lesions, suppression of immune surveillance, and tumor escape, more prone to leukemic evolution. Abstract Myelodysplasias with hypocellular bone marrow (hMDS) represent about 10–15% of MDS and are defined by reduced bone marrow cellularity (i.e., <25% or an inappropriately reduced cellularity for their age in young patients). Their diagnosis is still an object of debate and has not been clearly established in the recent WHO classification. Clinical and morphological overlaps with both normo/hypercellular MDS and aplastic anemia include cytopenias, the presence of marrow hypocellularity and dysplasia, and cytogenetic and molecular alterations. Activation of the immune system against the hematopoietic precursors, typical of aplastic anemia, is reckoned even in hMDS and may account for the response to immunosuppressive treatment. Finally, the hMDS outcome seems more favorable than that of normo/hypercellular MDS patients. In this review, we analyze the available literature on hMDS, focusing on clinical, immunological, and molecular features. We show that hMDS pathogenesis and clinical presentation are peculiar, albeit in-between aplastic anemia (AA) and normo/hypercellular MDS. Two different hMDS phenotypes may be encountered: one featured by inflammation and immune activation, with increased cytotoxic T cells, increased T and B regulatory cells, and better response to immunosuppression; and the other, resembling MDS, where T and B regulatory/suppressor cells prevail, leading to genetic clonal selection and an increased risk of leukemic evolution. The identification of the prevailing hMDS phenotype might assist treatment choice, inform prognosis, and suggest personalized monitoring.
Collapse
|
16
|
Awada H, Thapa B, Visconte V. The Genomics of Myelodysplastic Syndromes: Origins of Disease Evolution, Biological Pathways, and Prognostic Implications. Cells 2020; 9:E2512. [PMID: 33233642 PMCID: PMC7699752 DOI: 10.3390/cells9112512] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
The molecular pathogenesis of myelodysplastic syndrome (MDS) is complex due to the high rate of genomic heterogeneity. Significant advances have been made in the last decade which elucidated the landscape of molecular alterations (cytogenetic abnormalities, gene mutations) in MDS. Seminal experimental studies have clarified the role of diverse gene mutations in the context of disease phenotypes, but the lack of faithful murine models and/or cell lines spontaneously carrying certain gene mutations have hampered the knowledge on how and why specific pathways are associated with MDS pathogenesis. Here, we summarize the genomics of MDS and provide an overview on the deregulation of pathways and the latest molecular targeted therapeutics.
Collapse
Affiliation(s)
- Hassan Awada
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44106, USA;
| | - Bicky Thapa
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Valeria Visconte
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44106, USA;
| |
Collapse
|
17
|
Oka S, Ono K, Nohgawa M. The acquisition of trisomy 8 associated with Behçet's-like disease in myelodysplastic syndrome. Leuk Res Rep 2020; 13:100196. [PMID: 32211288 PMCID: PMC7082593 DOI: 10.1016/j.lrr.2020.100196] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/26/2020] [Accepted: 03/07/2020] [Indexed: 01/22/2023] Open
Abstract
A relationship has been reported between myelodysplastic syndrome (MDS) and autoimmune disease. Behçet's disease is a multisystem inflammatory disorder with mucocutaneous, articular, gastrointestinal, neurological, and vascular manifestations. The co-occurrence of MDS with trisomy 8 and Behçet's-like disease was recently demonstrated. We herein describe a case that shows the relationship between the acquisition of trisomy 8 and occurrence of Behçet's-like disease. Immune dysregulation and altered T-cell hemostasis play an important role in the pathogenesis of Behçet's-like disease and MDS with trisomy 8.
Collapse
Affiliation(s)
- Satoko Oka
- Division of Hematology, Japanese Red Cross Society Wakayama Medical Center, Wakayama, Japan
| | - Kazuo Ono
- Division of Pathology, Japanese Red Cross Society Wakayama Medical Center, Wakayama, Japan
| | - Masaharu Nohgawa
- Division of Hematology, Japanese Red Cross Society Wakayama Medical Center, Wakayama, Japan
| |
Collapse
|
18
|
Autoimmune disease in CMML-the chicken or the egg? Best Pract Res Clin Haematol 2019; 33:101136. [PMID: 32460986 DOI: 10.1016/j.beha.2019.101136] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 12/14/2022]
Abstract
Chronic myelomonocytic leukemia (CMML) is a clonal disorder that is associated with a wide range of systemic inflammatory and autoimmune diseases (SIADs). Approximately 20% of patients with CMML will have an associated SIAD and recognizing this association is critical to the evaluation, prognostication and management of patients with CMML. In this paper, we review the evidence supporting a causative link between these two entities as well as the direction of this relationship. We argue that the data favors CMML as the antecedent and causative disease state with a few notable exceptions. Better understanding of this relationship aids clinicians in the education of their patients and in determining the optimal management approach at the bedside. It is important to recognize opportunities to harmonize the treatments of these disease processes, which may enhance the effectiveness of treatment while reducing the burden of adverse effects from redundant therapies.
Collapse
|
19
|
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
|
20
|
Shallis RM, Ahmad R, Zeidan AM. Aplastic anemia: Etiology, molecular pathogenesis, and emerging concepts. Eur J Haematol 2018; 101:711-720. [PMID: 30055055 DOI: 10.1111/ejh.13153] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/23/2018] [Accepted: 07/24/2018] [Indexed: 12/12/2022]
Abstract
Aplastic anemia (AA) is rare disorder of bone marrow failure which if severe and not appropriately treated is highly fatal. AA is characterized by morphologic marrow features, namely hypocellularity, and resultant peripheral cytopenias. The molecular pathogenesis of AA is not fully understood, and a uniform process may not be the culprit across all cases. An antigen-driven and likely autoimmune dysregulated T-cell homeostasis is implicated in the hematopoietic stem cell injury which ultimately founds the pathologic features of the disease. Defective telomerase function and repair may also play a role in some cases as evidenced by recurring mutations in related telomerase complex genes such as TERT and TERC. In addition, recurring mutations in BCOR/BCORL, PIGA, DNMT3A, and ASXL1 as well as cytogenetic abnormalities, namely monosomy 7, trisomy 8, and uniparental disomy of the 6p arm seem to be intimately related to AA pathogenesis. The increased incidence of late clonal disease has also provided clues to accurately describe plausible predispositions to the development of AA. The emergence of newer genomic sequencing and other techniques is incrementally improving the understanding of the pathogenic mechanisms of AA, the detection of the disease, and ultimately offers the potential to improve patient outcomes. In this comprehensive review, we discuss the current understanding of the immunobiology, molecular pathogenesis, and future directions of such for AA.
Collapse
Affiliation(s)
- Rory M Shallis
- Division of Hematology/Medical Oncology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Rami Ahmad
- Division of Hematology/Medical Oncology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Amer M Zeidan
- Division of Hematology/Medical Oncology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut.,Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, Connecticut
| |
Collapse
|
21
|
Aanei CM, Catafal LC. Evaluation of bone marrow microenvironment could change how myelodysplastic syndromes are diagnosed and treated. Cytometry A 2018; 93:916-928. [PMID: 30211968 DOI: 10.1002/cyto.a.23506] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 05/06/2018] [Accepted: 05/17/2018] [Indexed: 12/13/2022]
Abstract
Myelodysplastic syndromes are a heterogeneous group of clonal hematopoietic disorders. However, the therapies used against the hematopoietic stem cells clones have limited efficacy; they slow the evolution toward acute myeloid leukemia rather than stop clonal evolution and eradicate the disease. The progress made in recent years regarding the role of the bone marrow microenvironment in disease evolution may contribute to progress in this area. This review presents the recent updates on the role of the bone marrow microenvironment in myelodysplastic syndromes pathogenesis and tries to find answers regarding how this information could improve myelodysplastic syndromes diagnosis and therapy.
Collapse
Affiliation(s)
- Carmen Mariana Aanei
- Laboratoire d'Hématologie, CHU de Saint-Etienne, 42055 Saint-Etienne Cedex 2, France
| | - Lydia Campos Catafal
- Laboratoire d'Hématologie, CHU de Saint-Etienne, 42055 Saint-Etienne Cedex 2, France
| |
Collapse
|
22
|
Rho H, Wells RA. A Game of Clones: The Complex Interplay of Aplastic Anaemia, Myelodysplastic Syndrome, and Paroxysmal Nocturnal Haemoglobinuria. EUROPEAN MEDICAL JOURNAL 2018. [DOI: 10.33590/emj/10312801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Although idiopathic aplastic anaemia (AA), myelodysplastic syndrome (MDS), and paroxysmal nocturnal haemoglobinuria (PNH) are all associated with bone marrow failure, they have traditionally been understood as distinct diseases with sharply contrasting pathophysiologies. These three disorders show considerable overlap. In 10% of cases of MDS the bone marrow is hypocellular, resembling AA, while glycophosphatidylinositol-deficient PNH cells can be detected in up to 5% of MDS and in >50% of AA patients. Results of recent studies offer a resolution to this overlap: MDS pathogenesis commonly has an autoimmune component and clonal haematopoiesis can be demonstrated in most cases of AA. Two explanations have arisen to explain the association of PNH with these disorders. It is hypothesised that PNH haematopoietic stem cells are relatively resistant to T cell attack and therefore have a competitive advantage in this context. Alternatively, it has been demonstrated that mutations associated with MDS are commonly present in PNH stem cells; such mutations could provide the PNH clone with an autonomous growth advantage. The authors suggest that these mechanisms may be necessary for the development of PNH in all cases, even when PNH occurs in the absence of MDS or AA. Finally, identification of a PNH clone is a predictive and prognostic biomarker in AA and MDS, adding important information for treatment and follow-up.
Collapse
Affiliation(s)
- Hayeong Rho
- Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Richard A Wells
- Faculty of Medicine, University of Toronto, Toronto, Canada; Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Canada
| |
Collapse
|
23
|
Shallis RM, Chokr N, Stahl M, Pine AB, Zeidan AM. Immunosuppressive therapy in myelodysplastic syndromes: a borrowed therapy in search of the right place. Expert Rev Hematol 2018; 11:715-726. [PMID: 30024293 DOI: 10.1080/17474086.2018.1503049] [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: 12/12/2022]
Abstract
INTRODUCTION Myelodysplastic syndromes (MDS) encompass a heterogenous collection of clonal hematopoietic stem cell disorders defined by dysregulated hematopoiesis, peripheral cytopenias, and a risk of leukemic progression. Increasing data support the role of innate and adaptive immune pathways in the pathogenesis and disease course of MDS. The role of immunosuppressive therapy has an established role in the treatment of other hematologic diseases, such as aplastic anemia whose pathogenesis is postulated to reflect that of MDS with regards to many aspects of immune activation. Areas covered: This paper discusses the current understanding of immune dysregulation as it pertains to MDS, the clinical experience with immunosuppressive therapy in the management of MDS, as well as future prospects which will likely improve therapeutic options and outcomes for patients with MDS. Expert commentary: Though limited by paucity of high quality data, immunomodulatory and immunosuppressive therapies for the treatment of MDS have shown meaningful clinical activity in selected patients. Continued clarification of the immune pathways that are dysregulated in MDS and establishing predictors for clinical benefit of immunosuppressive therapy are vital to improve the use and outcomes with these therapies.
Collapse
Affiliation(s)
- Rory M Shallis
- a Division of Hematology/Medical Oncology, Department of Medicine , Yale University School of Medicine , New Haven , USA
| | - Nora Chokr
- a Division of Hematology/Medical Oncology, Department of Medicine , Yale University School of Medicine , New Haven , USA
| | - Maximilian Stahl
- a Division of Hematology/Medical Oncology, Department of Medicine , Yale University School of Medicine , New Haven , USA
| | - Alexander B Pine
- a Division of Hematology/Medical Oncology, Department of Medicine , Yale University School of Medicine , New Haven , USA
| | - Amer M Zeidan
- a Division of Hematology/Medical Oncology, Department of Medicine , Yale University School of Medicine , New Haven , USA.,b Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center , Yale University , New Haven , USA
| |
Collapse
|
24
|
Outcomes and mutational analysis of patients with lower-risk non-del5q myelodysplastic syndrome treated with antithymocyte globulin with or without ciclosporine A. Leuk Res 2018; 71:67-74. [DOI: 10.1016/j.leukres.2018.05.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 05/15/2018] [Accepted: 05/22/2018] [Indexed: 01/28/2023]
|
25
|
Drevon L, Marceau A, Maarek O, Cuccuini W, Clappier E, Eclache V, Cluzeau T, Richez V, Berkaoui I, Dimicoli-Salazar S, Bidet A, Vial JP, Park S, Vieira Dos Santos C, Kaphan E, Berthon C, Stamatoullas A, Delhommeau F, Abermil N, Braun T, Sapena R, Lusina D, Renneville A, Adès L, Raynaud S, Fenaux P. Myelodysplastic syndrome (MDS) with isolated trisomy 8: a type of MDS frequently associated with myeloproliferative features? A report by the Groupe Francophone des Myélodysplasies. Br J Haematol 2018; 182:843-850. [DOI: 10.1111/bjh.15490] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/25/2018] [Indexed: 11/28/2022]
Affiliation(s)
- Louis Drevon
- Hôpital Saint-Louis; Assistance Publique Hôpitaux de Paris (APHP); University Paris Diderot (Paris 7); Paris France
| | - Alice Marceau
- Centre Hospitalier Régional Universitaire (CHRU) de Lille; Lille France
| | - Odile Maarek
- Hôpital Saint-Louis; Assistance Publique Hôpitaux de Paris (APHP); University Paris Diderot (Paris 7); Paris France
| | - Wendy Cuccuini
- Hôpital Saint-Louis; Assistance Publique Hôpitaux de Paris (APHP); University Paris Diderot (Paris 7); Paris France
| | - Emmanuelle Clappier
- Hôpital Saint-Louis; Assistance Publique Hôpitaux de Paris (APHP); University Paris Diderot (Paris 7); Paris France
| | | | - Thomas Cluzeau
- Centre Hospitalier Universitaire (CHU) de Nice; Nice France
| | | | - Inès Berkaoui
- Centre Hospitalier Universitaire (CHU) de Nice; Nice France
| | | | | | | | | | | | | | - Céline Berthon
- Centre Hospitalier Régional Universitaire (CHRU) de Lille; Lille France
| | | | - François Delhommeau
- Hôpital Saint-Antoine; APHP; University Pierre et Marie Curie (Paris 6); Paris France
| | - Nassera Abermil
- Hôpital Saint-Antoine; APHP; University Pierre et Marie Curie (Paris 6); Paris France
| | - Thorsten Braun
- Hôpital Avicenne; APHP; University Paris 13; Bobigny France
| | - Rosa Sapena
- GFM (Groupe Francophone des Myélodysplasies); Hôpital Saint-Louis; Paris France
| | - Daniel Lusina
- Hôpital Avicenne; APHP; University Paris 13; Bobigny France
| | - Aline Renneville
- Centre Hospitalier Régional Universitaire (CHRU) de Lille; Lille France
| | - Lionel Adès
- Hôpital Saint-Louis; Assistance Publique Hôpitaux de Paris (APHP); University Paris Diderot (Paris 7); Paris France
| | - Sophie Raynaud
- Centre Hospitalier Universitaire (CHU) de Nice; Nice France
| | - Pierre Fenaux
- Hôpital Saint-Louis; Assistance Publique Hôpitaux de Paris (APHP); University Paris Diderot (Paris 7); Paris France
| |
Collapse
|
26
|
Shallis RM, Ahmad R, Zeidan AM. The genetic and molecular pathogenesis of myelodysplastic syndromes. Eur J Haematol 2018; 101:260-271. [PMID: 29742289 DOI: 10.1111/ejh.13092] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2018] [Indexed: 12/14/2022]
Abstract
Myelodysplastic syndromes (MDS) comprise a diverse group of clonal and malignant myeloid disorders characterized by ineffective hematopoiesis, resultant peripheral cytopenias, and a meaningful increased risk of progression to acute myeloid leukemia. A wide array of recurring genetic mutations involved in RNA splicing, histone manipulation, DNA methylation, transcription factors, kinase signaling, DNA repair, cohesin proteins, and other signal transduction elements has been identified as important substrates for the development of MDS. Cytogenetic abnormalities, namely those characterized by loss of genetic material (including 5q- and 7q-), have also been strongly implicated and may influence the clonal architecture which predicts such mutations and may provoke an inflammatory bone marrow microenvironment as the substrate for clonal expansion. Other aspects of the molecular pathogenesis of MDS continue to be further elucidated, predicated upon advances in gene expression profiling and the development of new, and improved high-throughput techniques. More accurate understanding of the genetic and molecular basis for the development of MDS directly provides additional opportunity for treatment, which to date remains limited. In this comprehensive review, we examine the current understanding of the molecular pathogenesis and pathophysiology of MDS, as well as review future prospects which may enhance this understanding, treatment strategies, and hopefully outcomes.
Collapse
Affiliation(s)
- Rory M Shallis
- Division of Hematology/Medical Oncology, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Rami Ahmad
- Division of Hematology/Medical Oncology, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Amer M Zeidan
- Division of Hematology/Medical Oncology, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA.,Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT, USA
| |
Collapse
|
27
|
Zhang Y, Han S, Guo C, Zhang QX, Chang CK. [Effect of iron chelation therapy on EPO-Stat5 signaling pathway and Treg expression in IPSS low risk/medium risk-1 group myelodysplastic syndrome patients]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2018; 39:336-338. [PMID: 29779334 PMCID: PMC7342130 DOI: 10.3760/cma.j.issn.0253-2727.2018.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Indexed: 11/08/2022]
Affiliation(s)
| | | | | | | | - C K Chang
- Department of Hematology, The Sixth People's Hospital, Shanghai Jiaotong University, Shanghai 201306, China
| |
Collapse
|
28
|
|
29
|
Hemmati S, Haque T, Gritsman K. Inflammatory Signaling Pathways in Preleukemic and Leukemic Stem Cells. Front Oncol 2017; 7:265. [PMID: 29181334 PMCID: PMC5693908 DOI: 10.3389/fonc.2017.00265] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/20/2017] [Indexed: 12/15/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are a rare subset of bone marrow cells that usually exist in a quiescent state, only entering the cell cycle to replenish the blood compartment, thereby limiting the potential for errors in replication. Inflammatory signals that are released in response to environmental stressors, such as infection, trigger active cycling of HSCs. These inflammatory signals can also directly induce HSCs to release cytokines into the bone marrow environment, promoting myeloid differentiation. After stress myelopoiesis is triggered, HSCs require intracellular signaling programs to deactivate this response and return to steady state. Prolonged or excessive exposure to inflammatory cytokines, such as in prolonged infection or in chronic rheumatologic conditions, can lead to continued HSC cycling and eventual HSC loss. This promotes bone marrow failure, and can precipitate preleukemic states or leukemia through the acquisition of genetic and epigenetic changes in HSCs. This can occur through the initiation of clonal hematopoiesis, followed by the emergence preleukemic stem cells (pre-LSCs). In this review, we describe the roles of multiple inflammatory signaling pathways in the generation of pre-LSCs and in progression to myelodysplastic syndrome (MDS), myeloproliferative neoplasms, and acute myeloid leukemia (AML). In AML, activation of some inflammatory signaling pathways can promote the cycling and differentiation of LSCs, and this can be exploited therapeutically. We also discuss the therapeutic potential of modulating inflammatory signaling for the treatment of myeloid malignancies.
Collapse
Affiliation(s)
- Shayda Hemmati
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Tamanna Haque
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Oncology, Montefiore Medical Center, Bronx, NY, United States
| | - Kira Gritsman
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Oncology, Montefiore Medical Center, Bronx, NY, United States
| |
Collapse
|
30
|
Lenalidomide-mediated erythroid improvement in non-del(5q) myelodysplastic syndromes is associated with bone marrow immuno-remodeling. Leukemia 2017; 32:558-562. [PMID: 28972593 DOI: 10.1038/leu.2017.305] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
31
|
Zhang HF, Huang ZD, Wu XR, Li Q, Yu ZF. Comparison of T lymphocyte subsets in aplastic anemia and hypoplastic myelodysplastic syndromes. Life Sci 2017; 189:71-75. [PMID: 28935248 DOI: 10.1016/j.lfs.2017.09.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/14/2017] [Accepted: 09/18/2017] [Indexed: 12/26/2022]
Abstract
AIMS This study aims to compare the proportion of peripheral blood T lymphocyte subsets and related blood cell and bone marrow cytology indexes between patients with aplastic anemia (AA) and hypoplastic myelodysplastic syndrome (hypo-MDS), and investigate the clinical identification significance. MATERIALS AND METHODS A total of 41 patients with AA and 46 patients with hypo-MDS were collected, and the proportions of peripheral blood T lymphocyte subsets, CD3-CD16/CD56+NK cells, CD3+CD57+T-LGL cells and CD19+B lymphocytes were detected by flow cytometry. KEY FINDINGS The CD4+/CD8+ ratio decreased in the AA and hypo-MDS groups, and the difference was statistically significant when compared with that in the control group (P<0.05). However, there was no significant difference between AA and hypo-MDS groups (P>0.05). The proportions of CD3-CD16/CD56+NK cells and CD3+CD57+T-LGL cells in the hypo-MDS group were distinctly higher than those in the AA group (P<0.05). However, the proportion of CD19+B lymphocyte was obviously lower than that in the AA group (P<0.05). Furthermore, the proportions of reticulocytes, bone marrow progenitor cells and immature red blood cells in the hypo-MDS group were significantly more than those in the AA group (P<0.05), and the proportion of mature lymphocytes in the hypo-MDS group was distinctly lower than that in the AA group (P<0.05). SIGNIFICANCE Changes of T lymphocyte subsets and the proportions of large granular lymphocytes and B lymphocytes can be utilized as indexes in the differential diagnosis between AA and hypo-MDS.
Collapse
Affiliation(s)
- Hai-Fang Zhang
- Department of Chinese Medicine, School of Chinese Medicine Engineering, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, PR China
| | - Zhen-Dong Huang
- Department of Hematology, Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science, Tianjin 300020, PR China
| | - Xue-Run Wu
- Department of Chinese Medicine, School of Chinese Medicine Engineering, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, PR China
| | - Qian Li
- Department of Chinese Medicine, School of Chinese Medicine Engineering, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, PR China
| | - Zhi-Feng Yu
- Department of Chinese Medicine, School of Chinese Medicine Engineering, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, PR China.
| |
Collapse
|
32
|
List AF. Evolving Applications of Lenalidomide in the Management of Anemia in Myelodysplastic Syndromes. Cancer Control 2017; 13 Suppl:12-6. [PMID: 17242662 DOI: 10.1177/107327480601304s03] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Lenalidomide has emerged as an effective therapeutic alternative for the management of anemia in lower-risk myelodysplastic syndromes (MDS). Compelling results from phase I and phase II clinical studies prompted the US Food and Drug Administration to approve lenalidomide for the treatment of transfusion-dependent MDS patients with interstitial deletion of chromosome 5q [del(5q)]. Subsequently, the National Comprehensive Cancer Network (NCCN) has incorporated lenalidomide into their current treatment algorithm for the treatment of lower-risk del(5q) patients. This discussion examines the current NCCN guidelines for the treatment of these patients, including the management of anemia in lower-risk MDS, and discusses the potential future therapeutic applications of lenalidomide in this disease.
Collapse
Affiliation(s)
- Alan F List
- Malignant Hematology Program, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| |
Collapse
|
33
|
Cytogenetic Abnormalities in Myelodysplastic Syndromes: An Overview. Int J Hematol Oncol Stem Cell Res 2017; 11:231-239. [PMID: 28989590 PMCID: PMC5625474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Karyotype is one of the main constituents of the International Prognostic Scoring System (IPSS) and revised-IPSS that are the cornerstones for the prognostication of patients with myelodysplastic syndromes (MDS). Del(5q), -7/del(7q), +8 and -Y are among the most extensively studied cytogenetic abnormalities in MDS. The same applies for normal karyotype. There are hundreds of other rare cytogenetic abnormalities that have been reported in MDS, included but not limited to -X, 3q abnormalities, +13/del(13q), i(17q), +21/-21. However, due to a very low number of patients, their impact on the prognosis of MDS is limited. Knowledge of the molecular consequences of different cytogenetic abnormalities allows us to modify treatment regimens based on drugs most active against the specific karyotype present, allowing for the opportunity to individualize MDS treatment and improve patient care and prognosis.
Collapse
|
34
|
Barcellini W. The relationship between idiopathic cytopenias/dysplasias of uncertain significance (ICUS/IDUS) and autoimmunity. Expert Rev Hematol 2017; 10:649-657. [PMID: 28586251 DOI: 10.1080/17474086.2017.1339597] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION This review examines the several lines of evidence that support the relationship between myelodysplasia and autoimmunity, i.e. their epidemiologic association, the existence of common immune-mediated physiopathologic mechanisms, and the response to similar immunosuppressive therapies. The same relationship is reviewed here considering idiopathic cytopenia of uncertain significance (ICUS) and idiopathic dysplasia of uncertain significance (IDUS), two recently recognized provisional conditions characterized by isolated/unexplained cytopenia and/or dysplasia in <10% bone marrow cells. Areas covered: The review focuses on alterations of cytokine profiles, telomere/telomerase and toll-like receptors, and on increased myelosuppressive mediators and apoptotic markers in both myelodysplasia and autoimmunity. In addition, the presence of an autoimmune reaction directed against marrow precursors is described in refractory/relapsing autoimmune cytopenias (autoimmune hemolytic anemia, immune thrombocytopenia, chronic idiopathic neutropenia), possibly contributing to their evolution to ICUS/IDUS/bone marrow failure syndromes. Expert commentary: The increasing availability of omics methods has fuelled the discussion on the role of somatic mutations in the pathogenesis of IDUS/ICUS, clonal hematopoiesis of indeterminate potential, and clonal cytopenias of undetermined significance, and in their possible evolution. Even more attracting is the involvement of the genetic background/accumulating somatic mutations in cytopenias with autoimmune alterations.
Collapse
Affiliation(s)
- Wilma Barcellini
- a Onco-hematology Unit , IRCCS Ca' Granda - Maggiore Policlinico Hospital Foundation , Milan , Italy
| |
Collapse
|
35
|
Song Q, Peng M, Chu Y, Huang S. Techniques for detecting chromosomal aberrations in myelodysplastic syndromes. Oncotarget 2017; 8:62716-62729. [PMID: 28977983 PMCID: PMC5617543 DOI: 10.18632/oncotarget.17698] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 04/19/2017] [Indexed: 11/25/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are a group of heterogeneous hematologic diseases. Chromosomal aberrations are important for the initiation, development, and progression of MDS. Detection of chromosomal abnormalities in MDS is important for categorization, risk stratification, therapeutic selection, and prognosis evaluation of the disease. Recent progress of multiple techniques has brought powerful molecular cytogenetic information to reveal copy number variation, uniparental disomy, and complex chromosomal aberrations in MDS. In this review, we will introduce some common chromosomal aberrations in MDS and their clinical significance. Then we will explain the application, advantages, and limitations of different techniques for detecting chromosomal abnormalities in MDS. The information in this review may be helpful for clinicians to select appropriate methods in patient-related decision making.
Collapse
Affiliation(s)
- Qibin Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Min Peng
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuxin Chu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shiang Huang
- Molecular department, Kindstar Global, Wuhan, China
| |
Collapse
|
36
|
Stanley N, Olson TS, Babushok DV. Recent advances in understanding clonal haematopoiesis in aplastic anaemia. Br J Haematol 2017; 177:509-525. [PMID: 28107566 DOI: 10.1111/bjh.14510] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Acquired aplastic anaemia (AA) is an immune-mediated bone marrow failure disorder inextricably linked to clonal haematopoiesis. The majority of AA patients have somatic mutations and/or structural chromosomal abnormalities detected as early as at diagnosis. In contrast to other conditions linked to clonal haematopoiesis, the clonal signature of AA reflects its immune pathophysiology. The most common alterations are clonal expansions of cells lacking glycophosphotidylinositol-anchored proteins, loss of human leucocyte antigen alleles, and mutations in BCOR/BCORL1, ASXL1 and DNMT3A. Here, we present the current knowledge of clonal haematopoiesis in AA as it relates to aging, inherited bone marrow failure, and the grey-zone overlap of AA and myelodysplastic syndrome (MDS). We conclude by discussing the significance of clonal haematopoiesis both for improved diagnosis of AA, as well as for a more precise, personalized approach to prognostication of outcomes and therapy choices.
Collapse
Affiliation(s)
- Natasha Stanley
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Timothy S Olson
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Blood and Marrow Transplant Program, Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, USA
| | - Daria V Babushok
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Division of Hematology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
37
|
Kanamitsu K, Shimada A, Nishiuchi R, Shigemura T, Nakazawa Y, Koike K, Kodama Y, Shinkoda Y, Kawano Y, Yasui K, Sasaki K, Kajiwara R, Tsukahara H, Manabe A. Pediatric intestinal Behçet disease complicated by myeloid malignancies. Int J Hematol 2016; 105:377-382. [DOI: 10.1007/s12185-016-2127-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 11/01/2016] [Accepted: 11/02/2016] [Indexed: 02/04/2023]
|
38
|
Fozza C, Barraqueddu F, Corda G, Contini S, Virdis P, Dore F, Bonfigli S, Longinotti M. Study of the T-cell receptor repertoire by CDR3 spectratyping. J Immunol Methods 2016; 440:1-11. [PMID: 27823906 DOI: 10.1016/j.jim.2016.11.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 09/26/2016] [Accepted: 11/02/2016] [Indexed: 11/28/2022]
Abstract
The T-cell receptor (TCR) is the key player within the so called immunological synapse and the analysis of its repertoire offers a picture of both versatility and wideness of the whole immune T-cell compartment. Among the different approaches applied to its study the so-called spectratyping identifies the pattern of the third complementarity determining region (CDR3) length distribution in each one of the beta variable (TRBV) subfamilies encoded by the corresponding genes. This technique consists in a CDR3 fragment analysis through capillary electrophoresis, performed after cell separation, RNA extraction and reverse transcriptase PCR. This review will run through the most relevant studies which have tried to dissect the TCR repertoire usage in patients with different immune-mediated and infective diseases as well as solid or haematologic malignancies.
Collapse
Affiliation(s)
- Claudio Fozza
- Hematology, Department of Clinical and Experimental Medicine, University of Sassari, Viale San Pietro 12, 07100 Sassari, Italy.
| | - Francesca Barraqueddu
- Hematology, Department of Clinical and Experimental Medicine, University of Sassari, Viale San Pietro 12, 07100 Sassari, Italy
| | - Giovanna Corda
- Hematology, Department of Clinical and Experimental Medicine, University of Sassari, Viale San Pietro 12, 07100 Sassari, Italy
| | - Salvatore Contini
- Hematology, Department of Clinical and Experimental Medicine, University of Sassari, Viale San Pietro 12, 07100 Sassari, Italy
| | - Patrizia Virdis
- Hematology, Department of Clinical and Experimental Medicine, University of Sassari, Viale San Pietro 12, 07100 Sassari, Italy
| | - Fausto Dore
- Hematology, Department of Clinical and Experimental Medicine, University of Sassari, Viale San Pietro 12, 07100 Sassari, Italy
| | - Silvana Bonfigli
- Hematology, Department of Clinical and Experimental Medicine, University of Sassari, Viale San Pietro 12, 07100 Sassari, Italy
| | - Maurizio Longinotti
- Hematology, Department of Clinical and Experimental Medicine, University of Sassari, Viale San Pietro 12, 07100 Sassari, Italy
| |
Collapse
|
39
|
Fozza C, Crobu V, Isoni MA, Dore F. The immune landscape of myelodysplastic syndromes. Crit Rev Oncol Hematol 2016; 107:90-99. [PMID: 27823655 DOI: 10.1016/j.critrevonc.2016.08.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 08/15/2016] [Accepted: 08/31/2016] [Indexed: 12/25/2022] Open
Abstract
Even though the pathogenesis of myelodysplastic syndromes (MDS) is dominated by specific molecular defects involving hematopoietic precursors, also immune mechanisms seem to play a fundamental functional role. In this review we will first describe the clinical and laboratory autoimmune manifestations often detectable in MDS patients. We will then focus on studies addressing the possible influence of different immune cell subpopulations on the disease onset and evolution. We will finally consider therapeutic approaches based on immunomodulation, ranging from immunosuppressants to vaccination and transplantation strategies.
Collapse
Affiliation(s)
- Claudio Fozza
- Department of Clinical and Experimental Medicine, University of Sassari, Viale San Pietro 12, 07100 Sassari, Italy.
| | - Valeria Crobu
- Department of Clinical and Experimental Medicine, University of Sassari, Viale San Pietro 12, 07100 Sassari, Italy
| | - Maria Antonia Isoni
- Department of Clinical and Experimental Medicine, University of Sassari, Viale San Pietro 12, 07100 Sassari, Italy
| | - Fausto Dore
- Department of Clinical and Experimental Medicine, University of Sassari, Viale San Pietro 12, 07100 Sassari, Italy
| |
Collapse
|
40
|
Zeidan AM, Stahl M, Komrokji R. Emerging biological therapies for the treatment of myelodysplastic syndromes. Expert Opin Emerg Drugs 2016; 21:283-300. [DOI: 10.1080/14728214.2016.1220534] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
41
|
Kittang AO, Sand K, Brenner AK, Rye KP, Bruserud Ø. The Systemic Profile of Soluble Immune Mediators in Patients with Myelodysplastic Syndromes. Int J Mol Sci 2016; 17:ijms17071080. [PMID: 27399678 PMCID: PMC4964456 DOI: 10.3390/ijms17071080] [Citation(s) in RCA: 13] [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: 04/16/2016] [Revised: 06/20/2016] [Accepted: 06/30/2016] [Indexed: 01/20/2023] Open
Abstract
Introduction: Myelodysplastic syndromes (MDS) are characterized by bone marrow failure due to disturbed bone marrow maturation. MDS is associated with increased risk of transformation to acute myeloid leukemia (AML) and features of immunological dysregulation. Materials and methods: Serum levels of 47 soluble immune mediators were examined in samples derived from 49 MDS patients (35 low-risk and 14 high-risk) and 23 healthy adults. Our patients represent an unselected population-based cohort. The mediators included cytokines, soluble adhesion proteins, matrix metalloproteases, and tissue inhibitors of proteases. Levels were determined using Luminex assays. Patients were classified as low- and high-risk based on the international prognostic scoring system (IPSS) score. Results: When comparing the serum levels of single mediators the MDS patients showed a relatively wide variation range for several mediators compared with healthy adults, especially interleukin 6 (IL-6), IL-8/CXCL8, CCL3, and CCL4. The high-risk patients had lower levels of epidermal growth factor (EGF), cluster of differentiation 40 ligand (CD40L), CCL5, CCL11, CXCL5, matrix metalloproteinase 1 (MMP-1), MMP-9, and tissue inhibitor of metalloproteinases 2 (TIMP-2) compared with low-risk patients. Unsupervised hierarchical cluster analysis visualized marked serum mediator profile differences between MDS patients; based on this analysis three patient subsets could be identified. The healthy adults were also included in this analysis and, as expected, they formed their own separate cluster, except for one outlier. Both low- and high-risk patients showed considerable heterogeneity with regard to serum profile, and this heterogeneity seems stable over time (one year follow-up). Finally, very few mediators differed between low- and high-risk patients, but hierarchical clustering based both on all mediators, as well as five selected mediators (EGF, CCL11, TIMP-2, MMP-1, and MMP-9) identified subsets of patients with significantly increased frequency of high-risk disease (χ-square test p = 0.0158 and p = 0.0148).
Collapse
Affiliation(s)
- Astrid Olsnes Kittang
- Department of Clinical Science, University of Bergen, Bergen N-5021, Norway.
- Division for Hematology, Department of Medicine, Haukeland University Hospital, Bergen N-5021, Norway.
| | - Kristoffer Sand
- Department of Clinical Science, University of Bergen, Bergen N-5021, Norway.
| | | | - Kristin Paulsen Rye
- Department of Clinical Science, University of Bergen, Bergen N-5021, Norway.
| | - Øystein Bruserud
- Department of Clinical Science, University of Bergen, Bergen N-5021, Norway.
- Division for Hematology, Department of Medicine, Haukeland University Hospital, Bergen N-5021, Norway.
| |
Collapse
|
42
|
Wolach O, Stone R. Autoimmunity and Inflammation in Myelodysplastic Syndromes. Acta Haematol 2016; 136:108-17. [PMID: 27337745 DOI: 10.1159/000446062] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 04/06/2016] [Indexed: 01/28/2023]
Abstract
Autoimmune and inflammatory conditions (AICs) are encountered in up to 25% of patients with myelodysplastic syndromes (MDS). A wide range of AICs have been reported in association with MDS and can range from limited clinical manifestations to systemic diseases affecting multiple organs. Vasculitides, connective tissue diseases, and inflammatory arthritis are frequently reported in different studies; noninfectious fever and constitutional symptoms at presentation are common. Associations between AICs and specific MDS characteristics vary by study, but the available data suggest that AICs cluster more often in younger patients with higher-risk MDS. In general, AICs do not seem to confer worse survival, although certain AICs may be associated with adverse outcome (e.g. vasculitis) or progression of MDS (Sweet's syndrome). Nonetheless, these complications may have a significant impact on quality of life and affect the timing and type of MDS-directed therapy. The mainstay of management of these complications in the short term relies on immunosuppressive drugs. Increasing evidence suggests that hypomethylating agents may be effective in treating these complications and reduce steroid dependence. While the pathogenesis of AICs is incompletely understood, growing appreciation of cellular immune deregulation, cytokine hypersecretion, and the genetic heterogeneity underlying MDS may improve our understanding of common pathways linking MDS, inflammation, and autoimmunity.
Collapse
Affiliation(s)
- Ofir Wolach
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Mass., USA
| | | |
Collapse
|
43
|
Glenthøj A, Ørskov AD, Hansen JW, Hadrup SR, O'Connell C, Grønbæk K. Immune Mechanisms in Myelodysplastic Syndrome. Int J Mol Sci 2016; 17:ijms17060944. [PMID: 27314337 PMCID: PMC4926477 DOI: 10.3390/ijms17060944] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 05/31/2016] [Accepted: 06/08/2016] [Indexed: 12/12/2022] Open
Abstract
Myelodysplastic syndrome (MDS) is a spectrum of diseases, characterized by debilitating cytopenias and a propensity of developing acute myeloid leukemia. Comprehensive sequencing efforts have revealed a range of mutations characteristic, but not specific, of MDS. Epidemiologically, autoimmune diseases are common in patients with MDS, fueling hypotheses of common etiological mechanisms. Both innate and adaptive immune pathways are overly active in the hematopoietic niche of MDS. Although supportive care, growth factors, and hypomethylating agents are the mainstay of MDS treatment, some patients—especially younger low-risk patients with HLA-DR15 tissue type—demonstrate impressive response rates after immunosuppressive therapy. This is in contrast to higher-risk MDS patients, where several immune activating treatments, such as immune checkpoint inhibitors, are in the pipeline. Thus, the dual role of immune mechanisms in MDS is challenging, and rigorous translational studies are needed to establish the value of immune manipulation as a treatment of MDS.
Collapse
Affiliation(s)
- Andreas Glenthøj
- Epi-/Genome Laboratory, Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen 2100, Denmark.
| | - Andreas Due Ørskov
- Epi-/Genome Laboratory, Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen 2100, Denmark.
| | - Jakob Werner Hansen
- Epi-/Genome Laboratory, Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen 2100, Denmark.
| | - Sine Reker Hadrup
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Frederiksberg 1870, Denmark.
| | - Casey O'Connell
- Jane Anne Nohl Division of Hematology, USC Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA.
- Stand up to Cancer Epigenetics Dream Team, Van Andel Research Institute, Grand Rapids, MI 49503, USA.
| | - Kirsten Grønbæk
- Epi-/Genome Laboratory, Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen 2100, Denmark.
- Stand up to Cancer Epigenetics Dream Team, Van Andel Research Institute, Grand Rapids, MI 49503, USA.
| |
Collapse
|
44
|
Yue QF, Chen L, She XM, Hu B, Hu Y, Zou P, Liu XY. Clinical Prognostic Factors in 86 Chinese Patients with Primary Myelodysplastic Syndromes and Trisomy 8: A Single Institution Experience. Yonsei Med J 2016; 57:358-64. [PMID: 26847287 PMCID: PMC4740527 DOI: 10.3349/ymj.2016.57.2.358] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 12/01/2014] [Accepted: 01/08/2015] [Indexed: 11/27/2022] Open
Abstract
PURPOSE The objective was to determine the characteristics and prognostic factors of 86 Chinese patients with trisomy 8 aberrations and compare the prognostic value of International Prognostic System (IPSS) and Revised IPSS (IPSS-R) in this cohort. MATERIALS AND METHODS A total of 86 cases diagnosed with primary myelodysplastic syndromes (MDS) with isolated tr8 or with tr8 and other additional cytogenetic aberrations diagnosed and treated at the Union Hospital, Tongji Medical College of Huazhong University of Science and Technology between July 2002 and March 2013 were reviewed. RESULTS The median survival of the entire group was 23.0 months, and acute myeloid leukemia (AML) developed in 43% (37/86) patients within the follow up time. The univariate analysis revealed that overall survival (OS) was correlated with age, thrombocytopenia, absolute neutrophil count, marrow blasts, cytogenetic status and red blood cell transfusion at diagnosis, and the multivariate analysis revealed that age, marrow blasts, cytogenetic status and transfusion dependence were independent parameters for the OS. The cytogenetic complexity and marrow blasts had the strongest impact on the AML transformation by multivariate analysis. Comparing the two prognostic systems, both two systems could successfully discriminate risk groups for survival. IPSS-R was more refined than IPSS for predicting OS, but had no advantage in predicting the risk of AML development. CONCLUSION This study confirmed the influence of clinical factors on the prognosis of 86 Chinese MDS patients with trisomy 8. In addition, IPSS-R can further refine prognostic discrimination in the IPSS risk categories.
Collapse
Affiliation(s)
- Qing Fang Yue
- Department of Medical Oncology, ShaanXi Provincial People's Hospital, Xi'an, Shaanxi, P.R. China
| | - Lei Chen
- Department of Hematology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Xiao Mei She
- Department of Hematology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Bin Hu
- Department of Hematology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Yu Hu
- Department of Hematology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Ping Zou
- Department of Hematology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Xin Yue Liu
- Department of Hematology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China.
| |
Collapse
|
45
|
Higher Risk Myelodysplastic Syndromes in Patients with Well-Controlled HIV Infection: Clinical Features, Treatment, and Outcome. Case Rep Hematol 2016; 2016:8502641. [PMID: 26904323 PMCID: PMC4745308 DOI: 10.1155/2016/8502641] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/27/2015] [Indexed: 12/21/2022] Open
Abstract
Introduction. In advanced HIV prior to combination antiretroviral therapy (ART), dysplastic marrow changes occurred and resolved with ART. Few reports of myelodysplastic syndromes (MDS) in well-controlled HIV exist and management is undefined. Methods. Patients with well-controlled HIV and higher risk MDS were identified; characteristics, treatment, and outcomes were reviewed. Results. Of 292 MDS patients since 1996, 1 (0.3%) was HIV-positive. A 56-year-old woman presented with cytopenias. CD4 was 1310 cells/mL and HIV viral load <40 copies/mL. Bone marrow biopsy showed RCMD and karyotype included del(5q) and del(7q); IPSS was intermediate-2 risk. She received azacitidine at 75% dose. Cycle 2, at full dose, was complicated by marrow aplasia and possible AML; she elected palliation. Three additional HIV patients with higher risk MDS, aged 56-64, were identified from the literature. All had deletions involving chromosomes 5 and 7. MDS treatment of 2 was not reported and one received palliation; all died of AML. Conclusion. Four higher risk MDS in well-controlled HIV were below the median age of diagnosis for HIV-negative patients; all had adverse karyotype. This is the first report of an HIV patient receiving MDS treatment with azacitidine. Cytopenias were profound and dosing in HIV patients should be considered with caution.
Collapse
|
46
|
Li X, Shi J, Wang M, Nie N, Shao Y, Ge M, Huang J, Huang Z, Zhang J, Zheng Y. Cyclosporine Combined with Levamisole for Lower-Risk Myelodysplastic Syndromes. Acta Haematol 2015; 134:138-45. [PMID: 25925993 DOI: 10.1159/000370097] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 11/25/2014] [Indexed: 11/19/2022]
Abstract
Clinical and experimental evidence suggests an immune-mediated pathophysiology in subjects with lower-risk myelodysplastic syndromes (MDS) in whom immunosuppressive therapy may be effective. The novel immunosuppressive strategy of cyclosporine A (CsA) alternately combined with levamisole (LMS; CsA + LMS regimen) can dramatically improve the response rate and survival in aplastic anemia from those of our previous study. Herein, we retrospectively analyzed the data of 89 lower-risk MDS patients who received the CsA + LMS regimen. A total of 63 patients (70.8%) achieved either complete remission or hematological improvement at 4 months. Overall, 51, 41 and 19 patients had erythroid, platelet and neutrophil responses, respectively. Following the CsA + LMS regimen, 6 patients progressed to more advanced MDS at a median interval of 5 months (range, 3-42 months). The estimated 24-month progression-free survival was 82.2% (95% CI, 72.84-91.56) for all patients. Within the median follow-up of 18.5 months (range, 7.0-61.0), 6 patients died. In conclusion, the CsA + LMS regimen alleviated cytopenias and improved survival and freedom from evolution, suggesting that it could be reserved as an alternative choice for lower-risk MDS.
Collapse
Affiliation(s)
- Xingxin Li
- Severe Aplastic Anemia Studying Program, State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Cutting the cord from myelodysplastic syndromes: chronic myelomonocytic leukemia-specific biology and management strategies. Curr Opin Hematol 2015; 22:163-70. [PMID: 25575034 DOI: 10.1097/moh.0000000000000112] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE OF REVIEW Chronic myelomonocytic leukemia (CMML) is a troublesome hematologic malignancy characterized by peripheral blood monocytosis, marrow dysplasia, cytopenias, frequent extramedullary involvement by clonal cells, and a propensity for progression to acute myeloid leukemia. Although previously considered a subtype of the myelodysplastic syndromes (MDS), CMML is now recognized as a distinct entity with unique biologic and clinical features. This change has created a scientific and clinical research landscape that makes it difficult to discern CMML-specific validated conclusions versus speculative extrapolation from more general MDS data. RECENT FINDINGS Here, we review recent biologic observations that support the current CMML WHO classification, such as the high frequency of SRSF2 and ASXL1 mutations compared with MDS and critical dependence of CMML cells on granulocyte-macrophage colony-stimulating factor signaling. In addition, we discuss CMML-specific prognostic tools and therapeutic results of agents developed for MDS in patients with CMML. SUMMARY The present review focuses on evidence supporting CMML ontology and identifies key clinical differences in the management of CMML and that of MDS subtypes.
Collapse
|
48
|
Fozza C, Corda G, Barraqueddu F, Virdis P, Contini S, Galleu A, Isoni A, Dore F, Angelucci E, Longinotti M. Azacitidine improves the T-cell repertoire in patients with myelodysplastic syndromes and acute myeloid leukemia with multilineage dysplasia. Leuk Res 2015. [DOI: 10.1016/j.leukres.2015.06.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
49
|
Saumell S, Solé F, Arenillas L, Montoro J, Valcárcel D, Pedro C, Sanzo C, Luño E, Giménez T, Arnan M, Pomares H, De Paz R, Arrizabalaga B, Jerez A, Martínez AB, Sánchez-Castro J, Rodríguez-Gambarte JD, Raya JM, Ríos E, Rodríguez-Rivera M, Espinet B, Florensa L. Trisomy 8, a Cytogenetic Abnormality in Myelodysplastic Syndromes, Is Constitutional or Not? PLoS One 2015; 10:e0129375. [PMID: 26066831 PMCID: PMC4466575 DOI: 10.1371/journal.pone.0129375] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/07/2015] [Indexed: 02/02/2023] Open
Abstract
Isolated trisomy 8 is not considered presumptive evidence of myelodysplastic syndrome (MDS) in cases without minimal morphological criteria. One reason given is that trisomy 8 (+8) can be found as a constitutional mosaicism (cT8M). We tried to clarify the incidence of cT8M in myeloid neoplasms, specifically in MDS, and the diagnostic value of isolated +8 in MDS. Twenty-two MDS and 10 other myeloid neoplasms carrying +8 were studied. Trisomy 8 was determined in peripheral blood by conventional cytogenetics (CC) and on granulocytes, CD3+ lymphocytes and oral mucosa cells by fluorescence in situ hybridization (FISH). In peripheral blood CC, +8 was seen in 4/32 patients. By FISH, only one patient with chronic myelomonocytic leukemia showed +8 in all cell samples and was interpreted as a cT8M. In our series +8 was acquired in all MDS. Probably, once discarded cT8M by FISH from CD3+ lymphocytes and non-hematological cells, +8 should be considered with enough evidence to MDS.
Collapse
Affiliation(s)
- Sílvia Saumell
- Laboratori de Citologia Hematològica i Citogenètica Molecular, Servei de Patologia, Hospital del Mar, Barcelona, Spain
- GRETNHE, Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Department of Medicine, Medicine Faculty, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Francesc Solé
- Institut de Recerca Contra la Leucèmia Josep Carreras, Cytogenetics Platform, Badalona, Spain
| | - Leonor Arenillas
- Laboratori de Citologia Hematològica i Citogenètica Molecular, Servei de Patologia, Hospital del Mar, Barcelona, Spain
- GRETNHE, Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Julia Montoro
- Servicio de Hematología, Hospital Vall d’Hebrón, Barcelona, Spain
| | - David Valcárcel
- Servicio de Hematología, Hospital Vall d’Hebrón, Barcelona, Spain
| | - Carme Pedro
- GRETNHE, Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Servei de Hematologia Clínica, Hospital del Mar, Barcelona, Spain
| | - Carmen Sanzo
- Servicio de Hematología, Hospital Central de Asturias, Oviedo, Spain
| | - Elisa Luño
- Servicio de Hematología, Hospital Central de Asturias, Oviedo, Spain
| | - Teresa Giménez
- Servei d’Hematologia, Hospital Universitari Joan XXIII, Tarragona, Spain
| | - Montserrat Arnan
- Servei d’Hematologia, Hospital Duran i Reynals, Institut Català d’Oncologia, L’Hospitalet del Llobregat, Spain
| | - Helena Pomares
- Servei d’Hematologia, Hospital Duran i Reynals, Institut Català d’Oncologia, L’Hospitalet del Llobregat, Spain
| | - Raquel De Paz
- Servicio de Hematología, Hospital Universitario de La Paz, Madrid, Spain
| | | | - Andrés Jerez
- Servicio de Hematología, Hospital Morales Meseguer, Murcia, Spain
| | - Ana B. Martínez
- Servicio de Hematología, Hospital Morales Meseguer, Murcia, Spain
| | | | | | - José M. Raya
- Servicio de Hematología, Hospital Universitario de Canarias, La Laguna,Tenerife, Spain
| | - Eduardo Ríos
- Sevicio de Hematologia, Hospital Universitario de Valme, Sevilla, Spain
| | - María Rodríguez-Rivera
- Laboratori de Citologia Hematològica i Citogenètica Molecular, Servei de Patologia, Hospital del Mar, Barcelona, Spain
- GRETNHE, Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Blanca Espinet
- Laboratori de Citologia Hematològica i Citogenètica Molecular, Servei de Patologia, Hospital del Mar, Barcelona, Spain
- GRETNHE, Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Lourdes Florensa
- Laboratori de Citologia Hematològica i Citogenètica Molecular, Servei de Patologia, Hospital del Mar, Barcelona, Spain
- GRETNHE, Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- * E-mail:
| |
Collapse
|
50
|
Kulasekararaj AG, Kordasti S, Basu T, Salisbury JR, Mufti GJ, du Vivier AWP. Chronic relapsing remitting Sweet syndrome--a harbinger of myelodysplastic syndrome. Br J Haematol 2015; 170:649-56. [PMID: 25962438 DOI: 10.1111/bjh.13485] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 03/24/2015] [Indexed: 12/13/2022]
Abstract
Sweet syndrome (SS) is an acute febrile neutrophilic dermatosis. It has been associated with malignant disease, especially acute myeloid leukaemia (AML), infections, autoimmune disorders and drugs, particularly granulocyte colony-stimulating factor (GCSF). No cause is found in the rest, which are labelled idiopathic. We describe 15 patients with SS, which we believe represent 'immune dysregulation' secondary to myelodysplastic syndrome (MDS). We initially identified 31 patients with SS in a cohort of 744 patients with MDS and 215 with AML seen over a 6-year period (2004-10). The cause in 16 patients could be attributed either to administration of GCSF or chemotherapy. The eruption was brief and resolved spontaneously or following withdrawal of GCSF. Fifteen patients however, had a chronic debilitating illness dominated by the skin eruptions. Diagnosis of chronic relapsing SS was delayed because the pathology was not always typical of classical neutrophil-rich SS and included lymphocytic and histiocytoid infiltrates and bone marrow was not always performed because the relevance of the eruption to MDS was often not immediately appreciated. All these patients had 'low risk' MDS, diagnosed at a median of 17 months (range 0-157) following the diagnosis of SS. We describe a chronic debilitating episodic clinically distinctive skin eruption with features of SS but not always definitive histopathology often associated with immunological abnormalities affecting other systems related to underlying low risk MDS.
Collapse
Affiliation(s)
- Austin G Kulasekararaj
- King's College London School of Medicine, Department of Haematological Medicine, London, UK.,Department of Haematology, King's College Hospital, London, UK
| | - Shahram Kordasti
- King's College London School of Medicine, Department of Haematological Medicine, London, UK.,Department of Haematology, King's College Hospital, London, UK
| | - Tanya Basu
- Department of Haematology, King's College Hospital, London, UK.,Department of Dermatology, King's College Hospital, London, UK
| | | | - Ghulam J Mufti
- King's College London School of Medicine, Department of Haematological Medicine, London, UK.,Department of Haematology, King's College Hospital, London, UK
| | | |
Collapse
|