1
|
Liu C, Zhang Y, Liao R, Wang L, Zhou X, Tan M, Xu K, Wang H, Wang Q, Zhao Y, Cui Z, Lan X. Single-cell RNA sequencing of bone marrow reveals the immune response mechanisms of lymphocytes under avian leukosis virus subgroup J infection. Poult Sci 2025; 104:104995. [PMID: 40121758 PMCID: PMC11981752 DOI: 10.1016/j.psj.2025.104995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 02/27/2025] [Accepted: 03/05/2025] [Indexed: 03/25/2025] Open
Abstract
Avian Leukosis Virus (ALV) can induce tumorigenesis and immune suppression by acting on lymphocytes in the bone marrow. In this study, single-cell RNA sequencing (scRNA-seq) was used to analyze chicken bone marrow lymphocytes under Avian Leukosis Virus subtype J (ALV-J) infection. Using subgroup-specific marker genes and cell state analysis, we identified 18 distinct cell clusters, including 8 T cell clusters, 2 B cell clusters, 5 tumor-like cell clusters, and 3 unidentified clusters. Gene expression analysis revealed that in the 10 T/B lymphocyte clusters, the differentially expressed genes in double-positive T cells, B1-like B cells, and cytotoxic T cells were highly enriched in pathways related to viral infection and immune response. These three cell populations exhibited high proportions and significant changes after infection, suggesting a strong immune response to ALV-J infection. Additionally, during ALV-J infection, the proportion of regulatory T cells and CTLA4 T cells increased, while immune suppressive factors TGFB1 and IL16 were highly expressed across the cell populations, indicating an immune-suppressive state in bone marrow lymphocytes. Moreover, ALV-J infected all cell populations; however, within the same cluster, only a fraction of the cells expressed ALV-J viral genes. Notably, in all cells expressing ALV-J viral genes, the "Rho family GTPase signaling pathway" associated with antiviral responses was activated. The Rho family, which is a key regulator of cytoskeletal reorganization and cell polarity, also plays a critical role in tumor cell proliferation and metastasis. Further analysis using Ingenuity Pathway Analysis (IPA) software predicted key upstream regulators of immune response, such as MYC and MCYN. In conclusion, this study identifies key genes and signaling pathways involved in immune responses of different lymphocyte subpopulations triggered by ALV-J infection in bone marrow. These findings contribute to a better understanding of the immune mechanisms in ALV-J-infected bone marrow lymphocytes and provide insights for discovering breeding loci for ALV-J resistance.
Collapse
Affiliation(s)
- Cheng Liu
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Yu Zhang
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Ruyu Liao
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Lecheng Wang
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Xinyi Zhou
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Min Tan
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Keyun Xu
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Haiwei Wang
- Chongqing Academy of Animal Science, Chongqing, China
| | - Qigui Wang
- Chongqing Academy of Animal Science, Chongqing, China
| | - Yongju Zhao
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Zhifu Cui
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Xi Lan
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| |
Collapse
|
2
|
Kawano Y, Kawano H, LaMere MW, LaMere EA, Byun DK, McGrath KE, Palis J, Bajaj J, Liesveld JL, Katayama Y, Yamazaki S, Kapur R, Calvi LM, Ho TC, Becker MW. IL-1R1 and IL-18 signals regulate mesenchymal stromal cells in an aged murine model of myelodysplastic syndromes. Blood 2025; 145:1632-1644. [PMID: 39841001 PMCID: PMC12000655 DOI: 10.1182/blood.2024024818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/23/2025] Open
Abstract
ABSTRACT Myelodysplastic syndromes (MDS) are age-related diseases characterized by bone marrow (BM) dysfunction and an increased risk for developing acute leukemia. Although there is growing evidence that highlight the crucial role of the BM microenvironment (BMME) in MDS, the specific influence of inflammation on BMME changes and the potential benefits of targeting cytokines therapeutically remain to be elucidated. We previously found that interleukin-1 (IL-1) is a driver of aging phenotypes of the BMME and hematopoietic stem and progenitor cells (HSPCs). In this study, BM samples from patients with MDS demonstrated upregulated levels of IL-1 family cytokines, including IL-18. Using highly purified primary BM-derived mesenchymal stromal cells (MSCs), both IL-1b and IL-18 were found to exert direct effects on MSCs, thus influencing their ability to support HSPCs and erythroid progenitors. This confirms the significant involvement of both these IL-1 family cytokines in regulating the BM niche. Furthermore, targeting IL-1 receptor type 1 mitigated these aging phenotypes in older mice. We subsequently employed an age-appropriate murine model of MDS by transplanting NUP98-HOXD13 transgenic mice (NHD13Tg) cells into aged wild-type mice. Treatment with inhibitors that targeted IL-1 receptor-associated kinase 4 (IRAK4) and NOD-like receptor family pyrin domain containing 3 (NLRP3) reversed the proliferation of dysfunctional MSCs and enhanced their functionality. In addition, IRAK4 inhibition selectively suppressed MDS clonal cells while sparing non-MDS cells in the BM. These findings suggest that targeting IL-1 signaling holds promise for MDS treatment by addressing the underlying myeloid malignancy and restoring the altered BMME via BM-MSCs.
Collapse
Affiliation(s)
- Yuko Kawano
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Division of Cell Regulation, Center of Experimental Medicine and System Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hiroki Kawano
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Division of Hematology/Oncology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Mark W. LaMere
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Division of Hematology/Oncology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Elizabeth A. LaMere
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Division of Hematology/Oncology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Daniel K. Byun
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Division of Hematology/Oncology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Kathleen E. McGrath
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY
| | - James Palis
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY
| | - Jeevisha Bajaj
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Department of Biomedical Genetics, University of Rochester, Rochester, NY
| | - Jane L. Liesveld
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Division of Hematology/Oncology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Yoshio Katayama
- Division of Hematology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Satoshi Yamazaki
- Division of Cell Regulation, Center of Experimental Medicine and System Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Reuben Kapur
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Laura M. Calvi
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Tzu-Chieh Ho
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Division of Hematology/Oncology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Michael W. Becker
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Division of Hematology/Oncology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| |
Collapse
|
3
|
Kynurenines as a Novel Target for the Treatment of Malignancies. Pharmaceuticals (Basel) 2021; 14:ph14070606. [PMID: 34201791 PMCID: PMC8308824 DOI: 10.3390/ph14070606] [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: 05/10/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 12/12/2022] Open
Abstract
Malignancies are unquestionably a significant public health problem. Their effective treatment is still a big challenge for modern medicine. Tumors have developed a wide range of mechanisms to evade an immune and therapeutic response. As a result, there is an unmet clinical need for research on solutions aimed at overcoming this problem. An accumulation of tryptophan metabolites belonging to the kynurenine pathway can enhance neoplastic progression because it causes the suppression of immune system response against cancer cells. They are also involved in the development of the mechanisms responsible for the resistance to antitumor therapy. Kynurenine belongs to the most potent immunosuppressive metabolites of this pathway and has a significant impact on the development of malignancies. This fact prompted researchers to assess whether targeting the enzymes responsible for its synthesis could be an effective therapeutic strategy for various cancers. To date, numerous studies, both preclinical and clinical, have been conducted on this topic, especially regarding the inhibition of indoleamine 2,3-dioxygenase activity and their results can be considered noteworthy. This review gathers and systematizes the knowledge about the role of the kynurenine pathway in neoplastic progression and the findings regarding the usefulness of modulating its activity in anticancer therapy.
Collapse
|
4
|
Fuchs O. Treatment of Lymphoid and Myeloid Malignancies by Immunomodulatory Drugs. Cardiovasc Hematol Disord Drug Targets 2019; 19:51-78. [PMID: 29788898 DOI: 10.2174/1871529x18666180522073855] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 05/05/2018] [Accepted: 05/14/2018] [Indexed: 06/08/2023]
Abstract
Thalidomide and its derivatives (lenalidomide, pomalidomide, avadomide, iberdomide hydrochoride, CC-885 and CC-90009) form the family of immunomodulatory drugs (IMiDs). Lenalidomide (CC5013, Revlimid®) was approved by the US FDA and the EMA for the treatment of multiple myeloma (MM) patients, low or intermediate-1 risk transfusion-dependent myelodysplastic syndrome (MDS) with chromosome 5q deletion [del(5q)] and relapsed and/or refractory mantle cell lymphoma following bortezomib. Lenalidomide has also been studied in clinical trials and has shown promising activity in chronic lymphocytic leukemia (CLL) and non-Hodgkin lymphoma (NHL). Lenalidomide has anti-inflammatory effects and inhibits angiogenesis. Pomalidomide (CC4047, Imnovid® [EU], Pomalyst® [USA]) was approved for advanced MM insensitive to bortezomib and lenalidomide. Other IMiDs are in phases 1 and 2 of clinical trials. Cereblon (CRBN) seems to have an important role in IMiDs action in both lymphoid and myeloid hematological malignancies. Cereblon acts as the substrate receptor of a cullin-4 really interesting new gene (RING) E3 ubiquitin ligase CRL4CRBN. This E3 ubiquitin ligase in the absence of lenalidomide ubiquitinates CRBN itself and the other components of CRL4CRBN complex. Presence of lenalidomide changes specificity of CRL4CRBN which ubiquitinates two transcription factors, IKZF1 (Ikaros) and IKZF3 (Aiolos), and casein kinase 1α (CK1α) and marks them for degradation in proteasomes. Both these transcription factors (IKZF1 and IKZF3) stimulate proliferation of MM cells and inhibit T cells. Low CRBN level was connected with insensitivity of MM cells to lenalidomide. Lenalidomide decreases expression of protein argonaute-2, which binds to cereblon. Argonaute-2 seems to be an important drug target against IMiDs resistance in MM cells. Lenalidomide decreases also basigin and monocarboxylate transporter 1 in MM cells. MM cells with low expression of Ikaros, Aiolos and basigin are more sensitive to lenalidomide treatment. The CK1α gene (CSNK1A1) is located on 5q32 in commonly deleted region (CDR) in del(5q) MDS. Inhibition of CK1α sensitizes del(5q) MDS cells to lenalidomide. CK1α mediates also survival of malignant plasma cells in MM. Though, inhibition of CK1α is a potential novel therapy not only in del(5q) MDS but also in MM. High level of full length CRBN mRNA in mononuclear cells of bone marrow and of peripheral blood seems to be necessary for successful therapy of del(5q) MDS with lenalidomide. While transfusion independence (TI) after lenalidomide treatment is more than 60% in MDS patients with del(5q), only 25% TI and substantially shorter duration of response with occurrence of neutropenia and thrombocytopenia were achieved in lower risk MDS patients with normal karyotype treated with lenalidomide. Shortage of the biomarkers for lenalidomide response in these MDS patients is the main problem up to now.
Collapse
Affiliation(s)
- Ota Fuchs
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague 2, Czech Republic
| |
Collapse
|
5
|
A Phase II Study to Determine the Safety and Efficacy of the Oral Inhibitor of Indoleamine 2,3-Dioxygenase (IDO) Enzyme INCB024360 in Patients with Myelodysplastic Syndromes. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2018; 19:157-161. [PMID: 30713125 DOI: 10.1016/j.clml.2018.12.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 12/11/2018] [Accepted: 12/12/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND INCB024360 is an oral inhibitor of the enzyme indoleamine 2,3-dioxygenase (IDO), which catalyzes the degradation of tryptophan to kynurenine. Preclinical data suggest that IDO1 inhibition by INCB024360 will increase T cell proliferation, and decrease T regulatory cells and myeloid derived suppressor cells suppressive activity. We conducted a phase II study to explore activity and pharmacodynamics of INCB024360 in patients with myelodysplastic syndromes. PATIENTS AND METHODS All patients were treated with INCB024360 600 mg orally twice a day for at least 16 weeks. Fifteen patients were enrolled. The median age was 72 years. The International Prognostic Scoring System risk was low in 27% (n = 4), intermediate-1 in 47% (n = 7), and intermediate-2 in 27% (n = 4). All patients had prior azacitidine. RESULTS The best response was stable disease in 12 (80%) patients and progressive disease in 3 (20%) patients. The treatment was relatively well-tolerated. One patient developed hypothyroidism and adrenal insufficiency (grade 2), and 1 patient had low testosterone level. The mean IDO expression was 39% at baseline and 26% after treatment (n = 9; P = .4). The mean burst forming unit-erythroid changed from 72 to 191 colonies/106 (n = 5; P = .036), and the mean colony forming unit-granulocye, monocyte from 62 to 180 colonies/106 (n = 6; P = .5). The mean myeloid derived suppressor cell % (CD33Lin-HLA cells) was 29.5% at baseline compared with 27.6% after treatment (n = 9; P = .7). The mean T-regulatory effector memory cell % changed from 9.6% at screening to 7.4% at end of treatment (n = 14; P = .8). The mean kynurenine/tryptophan ratio decreased from 45 at baseline to 26 (42% reduction) at cycle 2, day 1 (P < .005). CONCLUSION Future directions may include testing INCB024360 early in the course of the disease.
Collapse
|
6
|
Wang C, Yang Y, Gao S, Chen J, Yu J, Zhang H, Li M, Zhan X, Li W. Immune dysregulation in myelodysplastic syndrome: Clinical features, pathogenesis and therapeutic strategies. Crit Rev Oncol Hematol 2018; 122:123-132. [DOI: 10.1016/j.critrevonc.2017.12.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/26/2017] [Accepted: 12/18/2017] [Indexed: 12/16/2022] Open
|
7
|
Dong W, Ding T, Wu L, Ren X, Epling-Burnette PK, Yang L. Effect of IL-7 and IL-15 on T cell phenotype in myelodysplastic syndromes. Oncotarget 2018; 7:27479-88. [PMID: 27036031 PMCID: PMC5053665 DOI: 10.18632/oncotarget.8459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 03/16/2016] [Indexed: 11/25/2022] Open
Abstract
Aberrant T cell phenotype is one of the characteristics of myelodysplastic syndromes (MDS). In this study, we detected an increased concentration of IL-15 in the plasma of MDS patients (n = 20) compared with that in the plasma of healthy controls (n = 20). In MDS patients, reduced naïve CD4+ and CD8+ T cells [16.11 ± 6.56 vs. 24.11 ± 7.18 for CD4+ T cells (p < 0.001) and 13.15 ± 5.67 vs. 23.51 ± 6.25 for CD8+ T cells (p < 0.001)] were observed. The reduced naïve and increased effector memory T cells were significantly correlated with IL-15 plasma level. Then, the effect of IL-15 and IL-7 was tested in vitro. Peripheral blood mononuclear cells from MDS were treated for 15 days with IL-15. This treatment significantly decreased naïve CD4+ (p < 0.001) and CD8+ (p < 0.001) T cells and correspondingly increased terminal memory CD4+ and CD8+ T cells (p < 0.001). Treatment with IL-7 increased naïve CD4+ (p < 0.05) and CD8+ (p < 0.001) T cells. Our results indicated that exposure to high levels of IL-15 may be involved in the T cell phenotype conversion observed in MDS. IL-7 may be one of the promising therapeutic candidates for recovering the effector immune compartment in MDS patients.
Collapse
Affiliation(s)
- Wen Dong
- Department of Orthopaedic Surgery, Tianjin Hongqiao Hospital, Tianjin, P.R. China
| | - Tingting Ding
- Department of Immunology, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, P.R. China.,National Clinical Research Center of Cancer, P.R. China.,Tianjin Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P.R. China
| | - Lei Wu
- Department of Immunology, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, P.R. China.,National Clinical Research Center of Cancer, P.R. China.,Tianjin Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P.R. China
| | - Xiubao Ren
- Department of Immunology, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, P.R. China.,National Clinical Research Center of Cancer, P.R. China.,Tianjin Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P.R. China
| | | | - Lili Yang
- Department of Immunology, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, P.R. China.,National Clinical Research Center of Cancer, P.R. China.,Tianjin Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P.R. China
| |
Collapse
|
8
|
Li AJ, Calvi LM. The microenvironment in myelodysplastic syndromes: Niche-mediated disease initiation and progression. Exp Hematol 2017; 55:3-18. [PMID: 28826860 PMCID: PMC5737956 DOI: 10.1016/j.exphem.2017.08.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/10/2017] [Accepted: 08/11/2017] [Indexed: 01/23/2023]
Abstract
Myelodysplastic syndromes (MDSs) are clonal disorders of hematopoietic stem and progenitor cells and represent the most common cause of acquired marrow failure. Hallmarked by ineffective hematopoiesis, dysplastic marrow, and risk of transformation to acute leukemia, MDS remains a poorly treated disease. Although identification of hematopoietic aberrations in human MDS has contributed significantly to our understanding of MDS pathogenesis, evidence now identify the bone marrow microenvironment (BMME) as another key contributor to disease initiation and progression. With improved understanding of the BMME, we are beginning to refine the role of the hematopoietic niche in MDS. Despite genetic diversity in MDS, interaction between MDS and the BMME appears to be a common disease feature and therefore represents an appealing therapeutic target. Further understanding of the interdependent relationship between MDS and its niche is needed to delineate the mechanisms underlying hematopoietic failure and how the microenvironment can be targeted clinically. This review provides an overview of data from human MDS and murine models supporting a role for BMME dysfunction at several steps of disease pathogenesis. Although no models or human studies so far have combined all of these findings, we review current data identifying BMME involvement in each step of MDS pathogenesis organized to reflect the chronology of BMME contribution as the normal hematopoietic system becomes myelodysplastic and MDS progresses to marrow failure and transformation. Although microenvironmental heterogeneity and dysfunction certainly add complexity to this syndrome, data are already demonstrating that targeting microenvironmental signals may represent novel therapeutic strategies for MDS treatment.
Collapse
Affiliation(s)
- Allison J Li
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Laura M Calvi
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY.
| |
Collapse
|
9
|
A 4-lncRNA scoring system for prognostication of adult myelodysplastic syndromes. Blood Adv 2017; 1:1505-1516. [PMID: 29296792 DOI: 10.1182/bloodadvances.2017008284] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 07/06/2017] [Indexed: 12/21/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) not only participate in normal hematopoiesis but also contribute to the pathogenesis of acute leukemia. However, their clinical and prognostic relevance in myelodysplastic syndromes (MDSs) remains unclear to date. In this study, we profiled lncRNA expressions in 176 adult patients with primary MDS, and identified 4 lncRNAs whose expression levels were significantly associated with overall survival (OS). We then constructed a risk-scoring system with the weighted sum of these 4 lncRNAs. Higher lncRNA scores were associated with higher marrow blast percentages, higher-risk subtypes of MDSs (based on both the Revised International Prognostic Scoring System [IPSS-R] and World Health Organization classification), complex cytogenetic changes, and mutations in RUNX1, ASXL1, TP53, SRSF2, and ZRSR2, whereas they were inversely correlated with SF3B1 mutation. Patients with higher lncRNA scores had a significantly shorter OS and a higher 5-year leukemic transformation rate compared with those with lower scores. The prognostic significance of our 4-lncRNA risk score could be validated in an independent MDS cohort. In multivariate analysis, higher lncRNA scores remained an independent unfavorable risk factor for OS (relative risk, 4.783; P < .001) irrespective of age, cytogenetics, IPSS-R, and gene mutations. To our knowledge, this is the first report to provide a lncRNA platform for risk stratification of MDS patients. In conclusion, our integrated 4-lncRNA risk-scoring system is correlated with distinctive clinical and biological features in MDS patients, and serves as an independent prognostic factor for survival and leukemic transformation. This concise yet powerful lncRNA-based scoring system holds the potential to improve the current risk stratification of MDS patients.
Collapse
|
10
|
Lambert C, Wu Y, Aanei C. Bone Marrow Immunity and Myelodysplasia. Front Oncol 2016; 6:172. [PMID: 27489795 PMCID: PMC4953538 DOI: 10.3389/fonc.2016.00172] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 07/05/2016] [Indexed: 12/29/2022] Open
Abstract
Myelodysplastic syndrome (MDS) is characterized by an ineffective hematopoiesis with production of aberrant clones and a high cell apoptosis rate in bone marrow (BM). Macrophages are in charge of phagocytosis. Innate Immune cells and specific T cells are in charge of immunosurveillance. Little is known on BM cell recruitment and activity as BM aspirate is frequently contaminated with peripheral blood. But evidences suggest an active role of immune cells in protection against MDS and secondary leukemia. BM CD8+ CD28− CD57+ T cells are directly cytotoxic and have a distinct cytokine signature in MDS, producing TNF-α, IL-6, CCL3, CCL4, IL-1RA, TNFα, FAS-L, TRAIL, and so on. These tools promote apoptosis of aberrant cells. On the other hand, they also increase MDS-related cytopenia and myelofibrosis together with TGFβ. IL-32 produced by stromal cells amplifies NK cytotoxicity but also the vicious circle of TNFα production. Myeloid-derived suppressing cells (MDSC) are increased in MDS and have ambiguous role in protection/progression of the diseases. CD33 is expressed on hematopoietic stem cells on MDS and might be a potential target for biotherapy. MDS also has impact on immunity and can favor chronic inflammation and emergence of autoimmune disorders. BM is the site of hematopoiesis and thus contains a complex population of cells at different stages of differentiation from stem cells and early engaged precursors up to almost mature cells of each lineage including erythrocytes, megakaryocytes, myelo-monocytic cells (monocyte/macrophage and granulocytes), NK cells, and B cells. Monocytes and B cell finalize their maturation in peripheral tissues or lymph nodes after migration through the blood. On the other hand, T cells develop in thymus and are present in BM only as mature cells, just like other well vascularized tissues. BM precursors have a strong proliferative capacity, which is usually associated with a high risk for genetic errors, cell dysfunction, and consequent cell death. Abnormal cells are prone to destruction through spontaneous apoptosis or because of the immunosurveillance that needs to stay highly vigilant. High rates of proliferation or differentiation failures lead to a high rate of cell death and massive release of debris to be captured and destroyed (1). Numerous macrophages reside in BM in charge of home-keeping. They have a high capacity of phagocytosis required for clearing all these debris.
Collapse
Affiliation(s)
- Claude Lambert
- Immunology Laboratory, Pole de Biologie-Pathologie, University Hospital of St Etienne , St Etienne , France
| | - Yuenv Wu
- Haematology Laboratory, Pole de Biologie-Pathologie, University Hospital of St Etienne , St Etienne , France
| | - Carmen Aanei
- Haematology Laboratory, Pole de Biologie-Pathologie, University Hospital of St Etienne , St Etienne , France
| |
Collapse
|
11
|
Komrokji RS, Kulasekararaj A, Al Ali NH, Kordasti S, Bart-Smith E, Craig BM, Padron E, Zhang L, Lancet JE, Pinilla-Ibarz J, List AF, Mufti GJ, Epling-Burnette PK. Autoimmune diseases and myelodysplastic syndromes. Am J Hematol 2016; 91:E280-3. [PMID: 26875020 DOI: 10.1002/ajh.24333] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 02/10/2016] [Indexed: 12/19/2022]
Abstract
Immune dysregulation and altered T-cell hemostasis play important roles in the pathogenesis of myelodysplastic syndromes (MDS). Recent studies suggest an increased risk of MDS among patients with autoimmune diseases. Here, we investigated the prevalence of autoimmune diseases among MDS patients, comparing characteristics and outcomes in those with and without autoimmune diseases. From our study group of 1408 MDS patients, 391 (28%) had autoimmune disease, with hypothyroidism being the most common type, accounting for 44% (n = 171) of patients (12% among all MDS patients analyzed). Other autoimmune diseases with ≥5% prevalence included idiopathic thrombocytopenic purpura in 12% (n = 46), rheumatoid arthritis in 10% (n = 41), and psoriasis in 7% (n = 28) of patients. Autoimmune diseases were more common in female MDS patients, those with RA or RCMD WHO subtype, and those who were less dependent on red blood cell transfusion. Median overall survival (OS) was 60 months (95% CI, 50-70) for patients with autoimmune diseases versus 45 months (95% CI, 40-49) for those without (log-rank test, P = 0.006). By multivariate analysis adjusting for revised IPSS and age >60 years, autoimmune diseases were a statistically significant independent factor for OS (HR 0.78; 95% CI, 0.66-0.92; P = 0.004). The rate of acute myeloid leukemia (AML) transformation was 23% (n = 89) in MDS patients with autoimmune disease versus 30% (n = 301) in those without (P = 0.011). Patient groups did not differ in response to azacitidine or lenalidomide treatment. Autoimmune diseases are prevalent among MDS patients. MDS patients with autoimmune diseases have better OS and less AML transformation.
Collapse
Affiliation(s)
- Rami S. Komrokji
- Department of Malignant Hematology; H. Lee Moffitt Cancer Center and Research Institute; Tampa Florida
| | - Austin Kulasekararaj
- Department of Haematological Medicine; King's College Hospital; London United Kingdom
| | - Najla H. Al Ali
- Department of Malignant Hematology; H. Lee Moffitt Cancer Center and Research Institute; Tampa Florida
| | - Shahram Kordasti
- Department of Haematological Medicine; King's College Hospital; London United Kingdom
| | - Emily Bart-Smith
- Department of Haematological Medicine; King's College Hospital; London United Kingdom
| | - Benjamin M. Craig
- Department of Malignant Hematology; H. Lee Moffitt Cancer Center and Research Institute; Tampa Florida
| | - Eric Padron
- Department of Malignant Hematology; H. Lee Moffitt Cancer Center and Research Institute; Tampa Florida
| | - Ling Zhang
- Department of Malignant Hematology; H. Lee Moffitt Cancer Center and Research Institute; Tampa Florida
| | - Jeffrey E. Lancet
- Department of Malignant Hematology; H. Lee Moffitt Cancer Center and Research Institute; Tampa Florida
| | - Javier Pinilla-Ibarz
- Department of Malignant Hematology; H. Lee Moffitt Cancer Center and Research Institute; Tampa Florida
| | - Alan F. List
- Department of Malignant Hematology; H. Lee Moffitt Cancer Center and Research Institute; Tampa Florida
| | - Ghulam J. Mufti
- Department of Haematological Medicine; King's College Hospital; London United Kingdom
| | | |
Collapse
|
12
|
Dan C, Chi J, Wang L. Molecular mechanisms of the progression of myelodysplastic syndrome to secondary acute myeloid leukaemia and implication for therapy. Ann Med 2015; 47:209-17. [PMID: 25861829 DOI: 10.3109/07853890.2015.1009156] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Myelodysplastic syndrome (MDS) includes a heterogeneous group of clonal haematological stem cell disorders characterized by dysplasia, cytopenias, ineffective haematopoiesis, and an increased risk of progression to acute myeloid leukaemia (AML), which is also called secondary AML (sAML). Approximately one-third of patients with MDS will progress to sAML within a few months to a few years, and this type of transformation is more common and rapid in patients with high-risk MDS (HR-MDS). However, the precise mechanisms underlying the evolution of MDS to sAML remain unclear. Currently, chemotherapy for sAML has minimal efficacy. The only method of curing patients with sAML is allogeneic haematopoietic stem cell transplantation (Allo-HSCT). Unfortunately, only a few patients are appropriate for transplantation because this disease primarily affects older adult patients. Additionally, compared to de novo AML, sAML is more difficult to cure, and the prognosis is often worse. Therefore, it is important to clarify the molecular mechanisms of the progression of MDS to sAML and to explore the potent drugs for clinical use. This review will highlight several molecular mechanisms of the progression of MDS to sAML and new therapeutic strategies of this disease.
Collapse
Affiliation(s)
- Chunli Dan
- Department of Haematology, The First Affiliated Hospital of Chongqing Medical University , Chongqing , China
| | | | | |
Collapse
|
13
|
Balderman SR, Calvi LM. Biology of BM failure syndromes: role of microenvironment and niches. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2014; 2014:71-76. [PMID: 25696837 PMCID: PMC4419374 DOI: 10.1182/asheducation-2014.1.71] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The BM microenvironment and its components regulate hematopoietic stem and progenitor cell (HSC) fate. An abnormality in the BM microenvironment and specific dysfunction of the HSC niche could play a critical role in initiation, disease progression, and response to therapy of BM failure syndromes. Therefore, the identification of changes in the HSC niche in BM failure syndromes should lead to further knowledge of the signals that disrupt the normal microenvironment. In turn, niche disruption may contribute to disease morbidity, resulting in pancytopenia and clonal evolution, and its understanding could suggest new therapeutic targets for these conditions. In this chapter, we briefly review the evidence for the importance of the BM microenvironment as a regulator of normal hematopoiesis, summarize current knowledge regarding the role of dysfunctions in the BM microenvironment in BM failure syndromes, and propose a strategy through which niche stimulation can complement current treatment for myelodysplastic syndrome.
Collapse
Affiliation(s)
- Sophia R. Balderman
- Instructor of Medicine, Division of Hematology/Oncology, Wilmot Cancer Center, University of Rochester School of Medicine
| | - Laura M. Calvi
- Professor of Medicine, Pharmacology and Physiology, Neurologic Surgery, Wilmot Cancer Center, University of Rochester School of Medicine, 601 Elmwood Avenue Box 693, Rochester, NY 14642, (585) 275-5011
| |
Collapse
|
14
|
Mailloux AW, Epling-Burnette PK. Effector memory regulatory T-cell expansion marks a pivotal point of immune escape in myelodysplastic syndromes. Oncoimmunology 2014; 2:e22654. [PMID: 23524348 PMCID: PMC3601152 DOI: 10.4161/onci.22654] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Myleodysplastic syndromes (MDS) are pre-malignant hematopoietic diseases that can progress to acute myeloid leukemia (AML) progression in conjunction with changes in immune function. In this model of leukemia evolution, the expansion of immunosuppressive regulatory T cells (Tregs) contributes to immune escape. Here, we discuss the importance of Treg-memory phenotype switching as a poor prognostic indicator in MDS.
Collapse
Affiliation(s)
- Adam W Mailloux
- Immunology Program; H. Lee Moffitt Cancer Center & Research Institute; Tampa, FL USA
| | | |
Collapse
|
15
|
Kazama H, Teramura M, Kurihara S, Yoshinaga K, Kato T, Motoji T. Peroxiredoxin 2 expression is increased in neutrophils of patients with refractory cytopenia with multilineage dysplasia. Br J Haematol 2014; 166:720-8. [DOI: 10.1111/bjh.12954] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 03/31/2014] [Indexed: 01/13/2023]
Affiliation(s)
- Hiroshi Kazama
- Department of Haematology; Tokyo Women's Medical University; Tokyo Japan
| | - Masanao Teramura
- Department of Haematology; Tokyo Women's Medical University; Tokyo Japan
| | - Sachiko Kurihara
- Department of Haematology; Tokyo Women's Medical University; Tokyo Japan
| | - Kentaro Yoshinaga
- Department of Haematology; Tokyo Women's Medical University; Tokyo Japan
| | - Tomohiro Kato
- Clinical Proteomics and Molecular Medicine; St. Marianna University Graduate School of Medicine; Kanagawa Japan
| | - Toshiko Motoji
- Department of Haematology; Tokyo Women's Medical University; Tokyo Japan
| |
Collapse
|
16
|
Merkel DG, Nagler A. Toward resolving the unsettled role of iron chelation therapy in myelodysplastic syndromes. Expert Rev Anticancer Ther 2014; 14:817-29. [PMID: 24641787 DOI: 10.1586/14737140.2014.896208] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Transfusion dependent low risk myelodysplastic syndromes (MDS) patients, eventually develop iron overload. Iron toxicity, via oxidative stress, can damage cellular components and impact organ function. In thalassemia major patients, iron chelation therapy lowered iron levels with recovery of cardiac and liver functions and significant improvement in survival. Several noncontrolled studies show inferior survival in MDS patients with iron overload, including an increase in transplant-related mortality and infection risk while iron chelation appears to improve survival in both lower risk MDS patients and in stem cell transplant settings. Collated data are presented on the pathophysiological impact of iron overload; measuring techniques and chelating agents' therapy positive impact on hematological status and overall survival are discussed. Although suggested by retrospective analyses, the lack of clear prospective data of the beneficial effects of iron chelation on morbidity and survival, the role of iron chelation therapy in MDS patients remains controversial.
Collapse
Affiliation(s)
- Drorit G Merkel
- Division of Hematology, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel
| | | |
Collapse
|
17
|
Yang L, Eksioglu EA, Wei S. hTERT deficiency in naïve T cells affects lymphocyte homeostasis in myelodysplastic syndrome patients. Oncoimmunology 2013; 2:e26329. [PMID: 24386611 PMCID: PMC3875656 DOI: 10.4161/onci.26329] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 08/31/2013] [Indexed: 01/14/2023] Open
Abstract
Myelodysplastic syndromes (MDSs) are hematopoietic stem cell disorders with a high potential to develop into acute myeloid leukemia (AML). We have recently demonstrated that naïve T cells, but not memory T cells, from MDS patients exhibit a pronounced deficiency in the mRNA coding for the catalytic subunit of telomerase (hTERT). We discuss the importance of this finding for lymphocytic homeostasis in MDS patients.
Collapse
Affiliation(s)
- Lili Yang
- Department of Immunology; Tianjin Cancer Institute and Hospital; Tianjin Medical University; Tianjin, China ; Key Laboratory of Cancer Immunology and Biotherapy; Tianjin, China
| | - Erika A Eksioglu
- Immunology Program at the H. Lee Moffitt Cancer Center; Tampa, FL USA
| | - Sheng Wei
- Immunology Program at the H. Lee Moffitt Cancer Center; Tampa, FL USA
| |
Collapse
|
18
|
Leitch HA, Buckstein R, Shamy A, Storring JM. The immunomodulatory agents lenalidomide and thalidomide for treatment of the myelodysplastic syndromes: A clinical practice guideline. Crit Rev Oncol Hematol 2013; 85:162-92. [DOI: 10.1016/j.critrevonc.2012.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 07/08/2012] [Accepted: 07/10/2012] [Indexed: 12/27/2022] Open
|
19
|
Epling-Burnette PK, McDaniel J, Wei S, List AF. Emerging immunosuppressive drugs in myelodysplastic syndromes. Expert Opin Emerg Drugs 2012; 17:519-41. [PMID: 23163589 DOI: 10.1517/14728214.2012.736487] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Myelodysplastic syndromes (MDS) are characterized by dysplastic morphologic features and ineffective hematopoiesis. Pathophysiological characteristics change over time making therapeutic development a major challenge. In early MDS, cytopenias arise or are exacerbated by humoral and cellular immune-mediators that suppress hematopoietic progenitor survival and alter the bone marrow microenvironment. AREAS COVERED In this review, current immunosuppressive regimens are described. To identify new therapies that may enhance immunosuppressive therapy (IST) response and identify pharmacodynamic biomarkers for patient selection, the inflammasome, cytokines, metabolic pathways and signaling events are described. EXPERT OPINION Agents with the potential to induce early, durable hematologic remissions are needed and many new immunosuppressive agents are available for investigation. An immune-mediated mechanism is likely to contribute to MDS early after diagnosis. New approaches that interfere with inflammatory pathways in the bone marrow microenvironment may move closer toward sustained disease control in MDS.
Collapse
Affiliation(s)
- Pearlie K Epling-Burnette
- H. Lee Moffitt Cancer Center & Research Institute, Immunology Department, SRB 23033, 12902 Magnolia Dr, Tampa, FL 33612, USA.
| | | | | | | |
Collapse
|
20
|
Komrokji RS, Corrales-Yepez M, Kharfan-Dabaja MA, Al Ali NH, Padron E, Rollison DE, Pinilla-Ibarz J, Zhang L, Epling-Burnette PK, Lancet JE, List AF. Hypoalbuminemia is an independent prognostic factor for overall survival in myelodysplastic syndromes. Am J Hematol 2012; 87:1006-9. [PMID: 23090887 DOI: 10.1002/ajh.23303] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 06/20/2012] [Accepted: 06/21/2012] [Indexed: 11/09/2022]
Abstract
We hypothesized that hypoalbuminemia is an independent prognostic factor in patients with myelodysplastic syndromes (MDS). We analyzed records of 767 patients treated at Moffitt Cancer Center between January 2001 and December 2009 to evaluate the relationship between serum albumin (SA) at the time of presentation and overall survival (OS). Patients (median age of 69 years) were stratified into three groups based on SA concentration (≤3.5, 3.6-4.0, and >4.0 g/dL). Two-thirds of the patients had low or intermediate-1 International Prognostic Scoring System (IPSS)-based risk for MDS. Median OS by SA concentration of ≤3.5, 3.6-4.0, and >4.0 g/dL was 11, 23, and 34 months, respectively (P < 0.005), whereas rate of acute myeloid leukemia progression was highest in patients with low SA (≤3.5 g/dL). The SA level offered prognostic discrimination for outcomes within the lower and higher IPSS risk groups, as well as with the MD Anderson risk model. In multivariable analysis, SA was a significant independent co-variate for OS after adjustment for IPSS, age, serum ferritin, and transfusion dependence (hazard ratio = 0.8; 95% CI 0.6-0.9; P = 0.004). Our findings indicate that hypoalbuminemia is an independent prognostic biomarker that may serve as a surrogate representative of disease biology or comorbidities in patients with MDS.
Collapse
Affiliation(s)
- Rami S Komrokji
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
The clinical implication of SRSF2 mutation in patients with myelodysplastic syndrome and its stability during disease evolution. Blood 2012; 120:3106-11. [PMID: 22932795 DOI: 10.1182/blood-2012-02-412296] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Recurrent somatic mutation of SRSF2, one of the RNA splicing machinery genes, has been identified in a substantial proportion of patients with myelodysplastic syndrome (MDS). However, the clinical and biologic characteristics of MDS with this mutation remain to be addressed. In this study, 34 (14.6%) of the 233 MDS patients were found to have SRSF2 mutation. SRSF2 mutation was closely associated with male sex (P = .001) and older age (P < .001). It occurred concurrently with at least 1 additional mutation in 29 patients (85.3%) and was closely associated with RUNX1, IDH2, and ASXL1 mutations (P = .004, P < .001, and P < .001, respectively). Patients with SRSF2 mutation had an inferior overall survival (P = .010), especially in the lower risk patients. Further exploration showed that the prognostic impact of SRSF2 mutation might be attributed to its close association with old age. Sequential analyses in 173 samples from 66 patients showed that all SRSF2-mutated patients retained their original mutations, whereas none of the SRSF2-wild patients acquired a novel mutation during disease evolution. In conclusion, SRSF2 mutation is associated with distinct clinical and biologic features in MDS patients. It is stable during the clinical course and may play little role in disease progression.
Collapse
|
22
|
A new recurrent chromosomal translocation t(3;11)(q13;q14) in myelodysplastic syndromes associated with overexpression of the ILDR1 gene. Leuk Res 2012; 36:852-6. [PMID: 22365942 DOI: 10.1016/j.leukres.2012.01.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 01/24/2012] [Accepted: 01/30/2012] [Indexed: 11/22/2022]
Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous group of diseases characterized by ineffective hematopoiesis and an increased risk of evolution to acute myeloid leukemia (AML). In this study, the combination of conventional cytogenetic, FISH studies and molecular techniques allowed us to unveil a novel recurrent t(3;11)(q13;q14) causing the overexpression of the immunoglobulin-like domain-containing receptor (ILDR1) gene. The analysis of gene expression was extended to Refractory Anemia (RA) and Refractory Anemia with excess blasts (RAEB) cases revealing ILDR1 overexpression in 36% of RAEB subgroup. The biological implications of the ILDR1 overexpression in MDS pathogenesis and its potential prognostic significance should be further investigated.
Collapse
|
23
|
Ornstein MC, Sekeres MA. Combination strategies in myelodysplastic syndromes. Int J Hematol 2012; 95:26-33. [PMID: 22218883 DOI: 10.1007/s12185-011-0987-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 12/01/2011] [Indexed: 11/30/2022]
Abstract
The myelodysplastic syndromes (MDS) consist of an array of clonal hematological malignancies resulting from disorders of pluripotent hematopoietic stem cells. MDS is associated with a poor overall prognosis and patients are categorized as higher risk and lower risk on the basis of the International Prognostic Scoring System. Currently, lenalidomide, azacitidine, and decitabine are the only three FDA-approved drugs for MDS. Traditional therapies for MDS involve the administration of single agents providing either supportive measures or disease-modifying effects directed to slowing progression to acute myeloid leukemia (AML) and improving survival. Recently, however, there has been increasing evidence suggesting that the combination of drugs with different mechanisms of action offers substantial benefit in the form of diminished side effects, improved overall survival, and delayed progression to AML. Multiple studies indicate that when compared with traditional monotherapies, combining various medications with non-overlapping mechanisms of action and toxicities may result in significant benefit for patients with MDS. A variety of combination therapies with growth factors, DNA methytransferase inhibitors, histone deacetylase inhibitors, and immunosuppressant treatments provide encouraging data indicating that the successful future of MDS treatment rests in the combination of multiple treatments modalities to achieve improved clinical outcomes.
Collapse
Affiliation(s)
- Moshe C Ornstein
- Leukemia Program, Department of Hematologic Oncology and Blood Disorders, Cleveland Clinic Taussig Cancer Institute, Desk R35, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | | |
Collapse
|
24
|
Myelodysplastic syndromes: revisiting the role of the bone marrow microenvironment in disease pathogenesis. Int J Hematol 2012; 95:17-25. [DOI: 10.1007/s12185-011-1001-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 12/12/2011] [Indexed: 12/11/2022]
|
25
|
Komrokji RS, Sekeres MA, List AF. Management of lower-risk myelodysplastic syndromes: the art and evidence. Curr Hematol Malig Rep 2011; 6:145-53. [PMID: 21442178 DOI: 10.1007/s11899-011-0086-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Myelodysplastic syndromes (MDS) represent a spectrum of bone marrow failure with variable outcome. Patients with "lower-risk" disease have an expected median survival measured in years, and a low risk of leukemia progression. Patients with "higher-risk" MDS, on the other hand, have expected survival measured in months without treatment and rapid leukemia progression. The outcome of those distinct groups can be explained by different underlying disease biology. In clinical practice, patients are stratified into risk groups based on prognostic models, most commonly the International Prognostic Scoring System (IPSS). In higher-risk disease, the standard of care is hypomethylating agents to extend survival and suppress leukemia potential, and consideration of allogeneic stem cell transplantation, which remains the only curative option. Patients classified as having lower-risk disease begin treatment with management focused on ameliorating hematologic deficits, related symptoms, or both. This review of lower-risk MDS highlights the biology of the disease and models for risk stratification. We use a case-based format to discuss current options for treatment, including erythropoiesis-stimulating agents, hypomethylating agents, lenalidomide, immunosuppressive therapy, supportive care, and investigational agents.
Collapse
Affiliation(s)
- Rami S Komrokji
- H. Lee Moffitt Cancer Center and Research Institute, FOB-3rd Floor, 12902 Magnolia Drive, Tampa, FL 33612, USA.
| | | | | |
Collapse
|
26
|
&NA;. Use chelation therapy to reduce iron overload and improve survival in patients with myelodysplastic syndromes. DRUGS & THERAPY PERSPECTIVES 2011. [DOI: 10.2165/11207940-000000000-00000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
27
|
Ishibashi M, Tamura H, Ogata K. Disease progression mechanism in myelodysplastic syndromes: insight into the role of the microenvironment. Leuk Res 2011; 35:1449-52. [PMID: 21757231 DOI: 10.1016/j.leukres.2011.06.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2011] [Revised: 06/17/2011] [Accepted: 06/18/2011] [Indexed: 12/29/2022]
Abstract
The somatic mutation theory proposing that a sequential accumulation of genetic abnormalities plays a major role in cancer pathogenesis has not yet been confirmed for myelodysplastic syndromes (MDS). Meanwhile, recent data in some cancers has underscored the role of the microenvironment in tumor growth. MDS CD34+CD38- cells usually fail to repopulate after transplantation in mice, suggesting the importance of the microenvironment for MDS cells. Our recent data have provided a disease-progression model in which overproduction of interferon-γ and tumor necrosis factor-α in the microenvironment is the primary event. This causes B7-H1 molecule expression on MDS blasts, which generates a bifunctional signal inducing T-cell apoptosis and enhancing blast proliferation. The latter may provide more opportunity for developing secondary genetic changes.
Collapse
Affiliation(s)
- Mariko Ishibashi
- Division of Hematology, Department of Medicine, Nippon Medical School, Tokyo, Japan
| | | | | |
Collapse
|
28
|
Leitch HA. Optimizing therapy for iron overload in the myelodysplastic syndromes: recent developments. Drugs 2011; 71:155-77. [PMID: 21275444 DOI: 10.2165/11585280-000000000-00000] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The myelodysplastic syndromes (MDS) are characterized by cytopenias and risk of progression to acute myeloid leukaemia (AML). Most MDS patients eventually require transfusion of red blood cells for anaemia, placing them at risk of transfusional iron overload. In β-thalassaemia major, transfusional iron overload leads to organ dysfunction and death; however, with iron chelation therapy, organ function is improved, and survival improved to near normal and correlated with the degree of compliance with chelation. In lower-risk MDS, several nonrandomized studies suggest an adverse effect of iron overload on survival and that lowering iron with chelation may minimize this impact. Emerging data indicate that chelation may improve organ function, particularly hepatic function, and a minority of patients may have improvement in cell counts and decreased transfusion requirements. While guidelines for MDS generally recommend chelation in selected lower-risk patients, data from nonrandomized trials suggest iron overload may impact adversely on the outcome of higher-risk MDS and stem cell transplantation (SCT). This effect may be due to increased transplant-related mortality, infection and AML progression, and preliminary data suggest that lowering iron may be beneficial in this patient group. Other areas of active and future investigation include optimizing the monitoring of iron overload using imaging such as T2* MRI and measures of labile iron and oxidative stress; correlating new methods of measuring iron to clinical outcomes; clarifying the contribution of different cellular and extracellular iron pools to iron toxicity; optimizing chelation by using agents that access the appropriate iron pools to minimize the relevant clinical consequences in individual patients; and incorporating measures of quality of life and co-morbidities into clinical trials of chelation in MDS. It should be noted that chelation is costly and potentially toxic, and in MDS should be initiated after weighing potential risks and benefits for each patient until more definitive data are available. In this review, data on the impact of iron overload in MDS and SCT are discussed; for example, several noncontrolled studies show inferior survival in patients with iron overload in these clinical settings, including an increase in transplant-related mortality and infection risk. Possible mechanisms of iron toxicity include oxidative stress, which can damage cellular components, and the documented impact of lowering iron on organ function with measures such as iron chelation therapy includes an improvement in elevated liver transaminases. Lowering iron also appears to improve survival in both lower-risk MDS and SCT in nonrandomized studies. Selected aspects of iron metabolism, transport, storage and distribution that may be amenable to future intervention and improved removal of iron from important cellular sites are discussed, as are attempts to quantify quality of life and the importance of co-morbidities in measures to treat MDS, including chelation therapy.
Collapse
Affiliation(s)
- Heather A Leitch
- Division of Hematology, St Paul's Hospital and the University of British Columbia, 440–1144 Burrard Street, Vancouver, BC, Canada.
| |
Collapse
|
29
|
Abstract
The myelodysplastic syndromes (MDS) are a heterogeneous group of myeloid disorders characterised by impaired peripheral blood cell production due to bone marrow dysplasia affecting one or more of the major myeloid cell lines. MDS are one of five major categories of myeloid neoplasms according to the World Health Organization (WHO) classification system for haematological cancers. Given their cytological and cytogenetic heterogeneity, these diseases probably constitute a group of molecularly distinct entities with variable degrees of ineffective haematopoiesis and susceptibility to leukaemic transformation. Recent studies provide some insights into the physiopathology of MDS. In the early stages, one mechanism contributing to hypercellular marrow and peripheral blood cytopenia is a significant increase in programmed cell death (apoptosis) in haematopoietic cells. Furthermore, altered responses in relation to cytokines, the immune system and bone marrow stroma also contribute to the disease phenotype. Deletions of chromosome 5q31-q32 are the most common recurring cytogenetic abnormalities detected in MDS. The 5q- syndrome is a new entity recognised in the WHO classification since 2001 and is associated with a good prognosis. Haploinsufficiency of multiple genes mapping to the common deleted region at 5q31-32 may contribute to the pathogenesis of 5q- syndrome and other MDS with 5q- deletion. Many studies have demonstrated that altered DNA methylation and histone acetylation can alter gene transcription. Abnormal methylation of transcription promoter sites is universal in patients with MDS, and the number of involved loci is increased in high-risk disease and secondary leukaemias. A better understanding of the pathogenesis of MDS can contribute to the development of new treatments such as hypomethylating drugs, immunomodulatory agents such as lenalidomide, and immunosuppressive drugs aimed at reversing the specific alteration that results in improvement in patients with MDS.
Collapse
Affiliation(s)
- Mar Tormo
- Hematology and Oncology Service, Valencia University Clinic Hospital, Valencia, Spain.
| | | | | |
Collapse
|
30
|
Alimena G, Breccia M, Musto P, Cilloni D, D'Auria F, Latagliata R, Sanpaolo G, Gottardi E, Saglio G, Mandelli F. Erythroid response and decrease of WT1 expression after proteasome inhibition by bortezomib in myelodysplastic syndromes. Leuk Res 2010; 35:504-7. [PMID: 20971509 DOI: 10.1016/j.leukres.2010.08.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2010] [Revised: 08/17/2010] [Accepted: 08/28/2010] [Indexed: 01/09/2023]
Abstract
NF-kB is reported to be constitutively activated in a percentage of high-risk myelodysplastic syndrome carrying cytogenetic aberrations. Only few data are reported on the use of proteasome inhibitors in this subset of patients. We performed a study on efficacy and safety of bortezomib as a single agent in patients with myelodysplastic syndromes (MDS). Bortezomib was administered at 1.3mg/m(2) with a 1, 4, 8, 11-day schedule every 28 days, in 19 patients with IPSS low/intermediate 1 or intermediate2/high risk. Six out of 19 patients received all planned eight cycles. Hematologic toxicity was recorded in all patients, especially grade 3/4 neutropenia and grade 3/4 thrombocytopenia; non-hematologic side effects were recorded in 7 patients, but events were all of grade 1/2 toxicity. According to IWG 2006 criteria, 4 out of 19 patients (21%) achieved erythroid response and 9 patients (47%) showed stable disease. In patients with erythroid response bone marrow WT1 levels decreased from a median of 109 copies at baseline to a median of 14 copies at the end of treatment, whereas in patients with stable disease, median WT1 copies increased either in bone marrow and peripheral blood. In conclusion, bortezomib used alone in MDS shows modest hematologic efficacy but appears to affect the WT1 gene expression, which is typically increased in these diseases.
Collapse
Affiliation(s)
- Giuliana Alimena
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Via Benevento 6, 00161 Rome, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
|
32
|
Lenalidomide for treatment of myelodysplastic syndromes: current status and future directions. Hematol Oncol Clin North Am 2010; 24:377-88. [PMID: 20359632 DOI: 10.1016/j.hoc.2010.02.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Lenalidomide was approved by the US Food and Drug Administration (FDA) for treatment of transfusion-dependent lower-risk myelodysplastic syndrome patients with deletion (del) (5q) alone or with additional karyotype abnormalities. The approval was based on high rates of prolonged transfusion independence and complete cytogenetic response in this subset. In lower-risk non-del(5q) patients, meaningful erythroid responses also were reported with a low frequency of cytogenetic improvement, although inferior to that observed in the del(5q) patients. There is now a better understanding of the mechanism of the karyotype-dependent drug action, explaining the disparate response rates and frequency of myelosuppression. In del(5q) patients, lenalidomide suppresses the clone by inhibiting the nuclear sequestration of the haplodeficient cell cycle regulatory protein cdc25c, thereby promoting selective G2 arrest and apoptosis. In non-del(5q) patients, lenalidomide enhances erythropoietin receptor signaling. Future directions include use of biologic and molecular markers as predictive tools to select patients and use of combination strategies to overcome resistance to lenalidomide in del(5q) patients or enhance erythropoiesis in non-del 5 patients.
Collapse
|
33
|
Current world literature. Curr Opin Pediatr 2010; 22:117-26. [PMID: 20068414 DOI: 10.1097/mop.0b013e32833539b5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|