1
|
Yang J. Efficacy and safety of decitabine combined with arsenic trioxide in elderly high-risk myelodysplastic neoplasm patients: a retrospective study. Hematology 2025; 30:2485493. [PMID: 40167011 DOI: 10.1080/16078454.2025.2485493] [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: 11/20/2024] [Accepted: 03/24/2025] [Indexed: 04/02/2025] Open
Abstract
OBJECTIVES Elderly patients with high-risk myelodysplastic neoplasm (MDS) face poor outcomes with limited treatment options, often progressing to acute myeloid leukemia (AML). This study investigates the efficacy and safety of combining decitabine (DAC) with arsenic trioxide (ATO) as a novel therapeutic approach. METHODS A retrospective analysis was conducted on 120 elderly high-risk MDS patients, with 52 receiving ATO-DAC (ATO-DAC group) and 68 receiving DAC monotherapy (DAC group). Treatment outcomes were assessed through overall response rate (ORR), progression-free survival (PFS), and overall survival (OS). Adverse events were recorded and compared between the two groups. RESULTS The ATO-DAC group demonstrated a significantly higher ORR of 78.85% compared to 52.94% in the DAC group (P = 0.026). Median PFS was 7.5 months for the ATO-DAC group versus 5.0 months for the DAC group (P = 0.021), and median OS was 14.5 months compared to 11.5 months, respectively (P = 0.034). Although adverse events were more frequent in the ATO-DAC group, the safety profile remained manageable. These findings suggest that the ATO-DAC combination provides superior efficacy compared to DAC monotherapy. DISCUSSION The combination of DAC and ATO offers a promising and innovative treatment option for elderly high-risk MDS patients, enhancing response rates and survival outcomes while maintaining a manageable safety profile. CONCLUSION This study underscores the clinical relevance of this regimen, warranting further investigation in prospective trials.
Collapse
Affiliation(s)
- Jianzhong Yang
- Department of Hematology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian, People's Republic of China
| |
Collapse
|
2
|
Qiu H, Zhang C, Ma X, Li Y. Molecular insights and treatment innovations: Advancing outcomes in acute myeloid leukemia with myelodysplasia‑related changes (Review). Oncol Rep 2025; 53:54. [PMID: 40116086 DOI: 10.3892/or.2025.8887] [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: 11/05/2024] [Accepted: 02/10/2025] [Indexed: 03/23/2025] Open
Abstract
Acute myeloid leukemia, myelodysplasia‑related (AML‑MR), a challenging and aggressive subtype of AML, is characterized by unique genetic abnormalities and molecular features, which contribute to its poor prognosis compared with other AML subtypes. The present review summarizes the current understanding of AML‑MR pathogenesis, highlighting notable advancements in genetic and cytogenetic insights. Critical mutations, such as those in the tumor antigen p53 and additional sex combs like 1 genes, and their role in disease progression and resistance to treatment, are explored. The review further investigates how clonal evolution and cellular microenvironment alterations drive AML‑MR transformation and impact patient outcomes. Despite the poor outlook typically associated with AML‑MR, developments in treatment approaches offer hope. The present review considers the efficacy of novel therapeutic agents, including CPX‑351, hypomethylating agents and targeted molecular therapies. Additionally, innovations in immunotherapy and allogeneic hematopoietic stem cell transplantation are discussed as promising avenues to improve patient survival rates. The challenges of treating AML‑MR, particularly in elderly and pretreated patients, underline the necessity for individualized treatment strategies that consider both the biological complexity of the disease and the overall health profile of the patient. The present review focuses on the mechanisms of AML‑MR transformation, highlighting factors that may offer a crucial theoretical foundation and pave the way for future applications in precision medicine. Future research directions include exploring novel targeted therapies and combination regimens to mitigate the transformation risks and enhance the quality of life of patients with AML‑MR.
Collapse
Affiliation(s)
- Hong Qiu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Chaowei Zhang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Xiaochen Ma
- Department of Public Health, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Ying Li
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
3
|
Gagelmann N, Kröger N. Allogeneic Stem Cell Transplant for Myelofibrosis and Myelodysplastic Syndromes: A Contemporary Review. Am J Hematol 2025. [PMID: 40079242 DOI: 10.1002/ajh.27660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/31/2024] [Accepted: 02/27/2025] [Indexed: 03/15/2025]
Abstract
Allogeneic hematopoietic stem-cell transplantation (HCT) remains the only potentially curative therapy for patients with myelodysplastic neoplasms (MDS) and myelofibrosis (MF) and is the standard care for eligible patients with higher-risk disease. Despite significant advancements, both diseases pose unique challenges due to their clinical and molecular heterogeneity, necessitating personalized approaches to patient selection, timing, and transplant management. For MDS, genomic profiling has revolutionized prognostic frameworks such as IPSS-M, enabling tailored therapeutic decisions. In MF, driver mutations (e.g., JAK2, CALR, MPL) and additional high-risk molecular markers provide critical insights into disease biology and transplant outcomes. Optimal timing of HCT is critical, and recent models might help personalize treatment approaches. Molecular measurable residual disease monitoring has demonstrated prognostic value in both diseases, guiding preemptive strategies to mitigate relapse risk. Harnessing molecular technologies, clinical expertise, patient-centered decision-making, and innovative pharmaceutical strategies offers an exciting opportunity to shape a transformative and curative treatment framework. Here, we provide a contemporary review on HCT for MDS and MF, highlighting up-to-date insights into disease biology, standard of care, and recommendations, as well as open avenues.
Collapse
Affiliation(s)
- Nico Gagelmann
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
4
|
Teymouri F, Dasanu CA. Selecting optimal therapy for higher-risk myelodysplastic syndromes: present and future projections. Expert Opin Pharmacother 2025; 26:345-347. [PMID: 39956939 DOI: 10.1080/14656566.2025.2468467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Affiliation(s)
- Farzad Teymouri
- Department of Medicine, Eisenhower Health, Rancho Mirage, CA, USA
| | - Constantin A Dasanu
- Lucy Curci Cancer Center, Eisenhower Health, Rancho Mirage, CA, USA
- Department of Medical Oncology and Hematology, UC San Diego Health System, San Diego, CA, USA
| |
Collapse
|
5
|
Tasis A, Spyropoulos T, Mitroulis I. The Emerging Role of CD8 + T Cells in Shaping Treatment Outcomes of Patients with MDS and AML. Cancers (Basel) 2025; 17:749. [PMID: 40075597 PMCID: PMC11898900 DOI: 10.3390/cancers17050749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/13/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
CD8+ T cells are critical players in anti-tumor immunity against solid tumors, targeted by immunotherapies. Emerging evidence suggests that CD8+ T cells also play a crucial role in anti-tumor responses and determining treatment outcomes in hematologic malignancies like myelodysplastic neoplasms (MDS) and acute myeloid leukemia (AML). In this review, we focus on the implication of CD8+ T cells in the treatment response of patients with MDS and AML. First, we review reported studies of aberrant functionality and clonality of CD8+ T cells in MDS and AML, often driven by the immunosuppressive bone marrow microenvironment, which can hinder effective antitumor immunity. Additionally, we discuss the potential use of CD8+ T cell subpopulations, including memory and senescent-like subsets, as predictive biomarkers for treatment response to a variety of treatment regimens, such as hypomethylating agents, which is the standard of care for patients with higher-risk MDS, and chemotherapy which is the main treatment of patients with AML. Understanding the multifaceted role of CD8+ T cells and their interaction with malignant cells in MDS and AML will provide useful insights into their potential as prognostic/predictive biomarkers, but also uncover alternative approaches to novel treatment strategies that could reshape the therapeutic landscape, thus improving treatment efficacy, aiding in overcoming treatment resistance and improving patient survival in these challenging myeloid neoplasms.
Collapse
Affiliation(s)
- Athanasios Tasis
- Translational Research and Laboratory Medicine Unit, First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | - Theodoros Spyropoulos
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | - Ioannis Mitroulis
- Translational Research and Laboratory Medicine Unit, First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| |
Collapse
|
6
|
Nikolopoulos T, Bochalis E, Chatzilygeroudi T, Chondrou V, Dereki I, Athanasopoulou K, Zafeiropoulos J, Bourikas K, Patrinos GP, Symeonidis A, Sgourou A. Integrating advanced analytical methods to assess epigenetic marks affecting response to hypomethylating agents in higher risk myelodysplastic syndrome. Mol Med 2025; 31:59. [PMID: 39953389 PMCID: PMC11829487 DOI: 10.1186/s10020-025-01123-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 02/08/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Patients with higher-risk (HR) myelodysplastic syndrome (MDS), ineligible for allogeneic hematopoietic stem cell transplantation (alloHSCT), require prompt therapeutic interventions, such as treatment with hypomethylating agents (HMAs) to restore normal DNA methylation patterns, mainly of oncosuppressor genes, and consequently to delay disease progression and increase overall survival (OS). However, response assessment to HMA treatment relies on conventional methods with limited capacity to uncover a wide spectrum of underlying molecular events. METHODS We implemented liquid chromatography-tandem mass spectrometry (LC-MS/MS) to assess 5' methyl-2' deoxycytidine (5mdC), 5' hydroxy-methyl-2'-deoxycytidine (5hmdC) levels and global adenosine/thymidine ([dA]/[T]) ratio in bone marrow aspirates from twenty-one HR MDS patients, pre- and post-HMA treatment. Additionally, targeted methylation analysis was performed by interpretation of NGS-methylation (MeD-seq) data obtained from the same patient cohort. RESULTS LC/MS-MS analysis revealed a significant hypomethylation status in responders (Rs), already established at baseline and a trend for further DNA methylation reduction post-HMA treatment. Non-responders (NRs) reached statistical significance for DNA hypomethylation only post-HMA treatment. The 5hmdC epigenetic mark was approximately detected at 37.5-40% among NRs and Rs, implying the impairment of the natural active demethylation pathway, mediated by the ten-eleven (TET) 5mdC dioxygenases. R and NR subgroups displayed a [dA]/[T] ratio < 1 (0.727 - 0.633), supporting high frequences of 5mdC transition to thymidine. Response to treatment, according to whole genome MeD-seq data analysis, was associated with specific, scattered hypomethylated DMRs, rather than presenting a global effect across genome. MeD-seq analysis identified divergent epigenetic effects along chromosomes 7, 9, 12, 16, 18, 21, 22, X and Y. Within statistically significant selected chromosomal bins, genes encoding for proteins and non-coding RNAs with reversed methylation profiles between Rs and NRs, were highlighted. CONCLUSIONS Implementation of powerful analytical tools to identify the dynamic DNA methylation changes in HR MDS patients undergoing HMA therapy demonstrated that LC-MS/MS exerts high efficiency as a broad-based but rapid and cost-effective methodology (compared to MeD-seq) to decode different perspectives of the epigenetic background of HR MDS patients and possess discriminative efficacy of the response phenotype to HMA treatment.
Collapse
Affiliation(s)
- Theodoros Nikolopoulos
- Biology Laboratory, School of Science and Technology, Hellenic Open University, Patras, Greece
| | - Eleftherios Bochalis
- Biology Laboratory, School of Science and Technology, Hellenic Open University, Patras, Greece
| | - Theodora Chatzilygeroudi
- School of Health Sciences, Faculty of Medicine, Hematology Division, University of Patras, Patras, Greece
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Vasiliki Chondrou
- Biology Laboratory, School of Science and Technology, Hellenic Open University, Patras, Greece
| | - Irene Dereki
- Biology Laboratory, School of Science and Technology, Hellenic Open University, Patras, Greece
| | - Katerina Athanasopoulou
- Biology Laboratory, School of Science and Technology, Hellenic Open University, Patras, Greece
| | - John Zafeiropoulos
- Chemistry Laboratory, School of Science and Technology, Hellenic Open University, Patras, Greece
| | - Kyriakos Bourikas
- Chemistry Laboratory, School of Science and Technology, Hellenic Open University, Patras, Greece
| | - George P Patrinos
- Laboratory of Pharmacogenomics and Inaffiliationidualized Therapy, Department of Pharmacy, School of Health Sciences, University of Patras, University Campus, Rion, Patras, Greece
- College of Medicine and Health Sciences, Department of Genetics and Genomics, United Arab Emirates University, Al Ain, Abu Dhabi, UAE
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE
- Clinical Bioinformatics Unit, Department of Pathology, Faculty of Medicine and Health Sciences, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Argiris Symeonidis
- School of Health Sciences, Faculty of Medicine, Hematology Division, University of Patras, Patras, Greece.
| | - Argyro Sgourou
- Biology Laboratory, School of Science and Technology, Hellenic Open University, Patras, Greece.
| |
Collapse
|
7
|
Xue T, Tao Y, Wu Z, Yuan C, Wang Y, Xu H, Cai S, Wang Z, Lu J. Realgar induces apoptosis by inhibiting glycolysis via regulating STAT3 in myelodysplastic syndrome. JOURNAL OF ETHNOPHARMACOLOGY 2025; 341:119339. [PMID: 39788167 DOI: 10.1016/j.jep.2025.119339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 12/24/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Myelodysplastic syndrome (MDS) is a hematologic malignancy that presents a unique opportunity for traditional Chinese medicine (TCM) to demonstrate its distinctive value in treatment. Realgar, a component of TCM, has shown notable potential in alleviating clinical symptoms and improving the prognosis of MDS patients. However, the precise mechanisms underlying the treatment of MDS with realgar, particularly its effects on apoptosis-related pathways, remain poorly understood. AIM OF THE STUDY This study aimed to investigate the pro-apoptotic effects of realgar on MDS cells and to elucidate the underlying molecular mechanisms. MATERIALS AND METHODS We explored the targets and pathways of realgar's action on MDS using public databases, network pharmacology, and RNA sequencing. Various techniques were employed, including cell transfection, Cell Counting Kit-8 (CCK8) assay, Cellular Thermal Shift Assay (CETSA), Western blot (WB), quantitative real-time polymerase chain reaction (qRT-PCR), apoptosis and glycolysis assays, extracellular acidification rate (ECAR) and oxygen consumption rate (OCR) measurements, dual-luciferase reporter assays, and immunofluorescence, to investigate the regulatory mechanisms involving STAT3, glycolysis, and apoptosis. Hematoxylin and eosin (HE) staining was utilized to assess realgar's toxicity. Apoptosis and hemogram changes were analyzed to evaluate the therapeutic effect of realgar on MDS transgenic mice. RESULTS Analysis of public data indicated that apoptosis-related genes are downregulated in MDS patients. Through network pharmacology, CETSA, qRT-PCR, WB, apoptosis assays, and STAT3 overexpression cell transfection, we discovered that realgar inhibits STAT3 expression. Further investigation using RNA sequencing suggested that glycolysis may be involved in this regulatory process. ECAR, OCR, glycolysis assays, WB, apoptosis assays, and glycolysis inhibitor experiments demonstrated that glycolytic function was inhibited. Additionally, GLUT1 expression was significantly decreased, and GLUT1 was found to directly bind to STAT3. In MDS mice, realgar treatment enhanced levels of white blood cells, red blood cells, hemoglobin, and platelets, and increased apoptosis levels. CONCLUSION Our findings reveal that realgar exerts a significant pro-apoptotic effect on MDS cells in both in vivo and in vitro models. Further analysis demonstrated that realgar regulates the STAT3 pathway, leading to GLUT1-mediated glycolysis alterations that ultimately induce apoptotic pathways, as represented by BCL2. These discoveries hold significant implications for the basic research and clinical diagnosis and treatment of MDS.
Collapse
Affiliation(s)
- Tingting Xue
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuchen Tao
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zong Wu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenyue Yuan
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanlu Wang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hao Xu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuyang Cai
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Ziliang Wang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Jiahui Lu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
8
|
Teymouri F, Dasanu CA. Selecting appropriate therapy for lower-risk myelodysplastic syndromes: current state and future prospects. Expert Opin Pharmacother 2024; 25:1975-1977. [PMID: 39387446 DOI: 10.1080/14656566.2024.2415714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 10/15/2024]
Affiliation(s)
- Farzad Teymouri
- Department of Medicine, Eisenhower Health, Rancho Mirage, CA, USA
| | - Constantin A Dasanu
- Lucy Curci Cancer Center, Eisenhower Health, Rancho Mirage, CA, USA
- Department of Medical Oncology and Hematology, UC San Diego Health System, San Diego, CA, USA
| |
Collapse
|
9
|
Battaglia MR, Cannova J, Madero-Marroquin R, Patel AA. Treatment of Anemia in Lower-Risk Myelodysplastic Syndrome. Curr Treat Options Oncol 2024; 25:752-768. [PMID: 38814537 DOI: 10.1007/s11864-024-01217-0] [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] [Accepted: 05/13/2024] [Indexed: 05/31/2024]
Abstract
OPINION STATEMENT A majority of patients with lower-risk myelodysplastic syndrome (MDS) will present with or develop anemia. Anemia in MDS is associated with decreased quality of life and may correlate with decreased progression-free survival and overall survival. In this state of the art review we summarize current risk stratification approaches to identify lower-risk MDS (LR-MDS), the natural history of the disease, and meaningful clinical endpoints. The treatment landscape of LR-MDS with anemia is also rapidly evolving; we review the role of supportive care, erythropoietin stimulating agents, lenalidomide, luspatercept, hypomethylating agents (HMAs), and immunosuppressive therapy (IST) in the management of LR-MDS with anemia. In patients with deletion 5q (del5q) syndrome lenalidomide has both efficacy and durability of response. For patients without del5q who need treatment, the management approach is impacted by serum erythropoietin (EPO) level, SF3B1 mutation status, and ring sideroblast status. Given the data from the Phase III COMMANDS trial, we utilize luspatercept in those with SF3B1 mutation or ring sideroblasts that have an EPO level < 500 U/L; in patients without an SF3B1 mutation or ring sideroblasts there is equipoise between luspatercept and use of an erythropoietin stimulating agent (ESA). For patients who have an EPO level ≥ 500 U/L or have been previously treated there is not a clear standard of care. For those without previous luspatercept exposure it can be considered particularly if there is an SF3B1 mutation or the presence of ring sideroblasts. Other options include HMAs or IST; the Phase III IMERGE trial supports the efficacy of the telomerase inhibitor imetelstat in this setting and this may become a standard option in the future as well.
Collapse
Affiliation(s)
| | - Joseph Cannova
- Section of Hematology-Oncology, Department of Medicine, University of Chicago, 5841 S Maryland Avenue MC 2115, Chicago, IL, 60637, USA
| | - Rafael Madero-Marroquin
- Section of Hematology-Oncology, Department of Medicine, University of Chicago, 5841 S Maryland Avenue MC 2115, Chicago, IL, 60637, USA
| | - Anand A Patel
- Section of Hematology-Oncology, Department of Medicine, University of Chicago, 5841 S Maryland Avenue MC 2115, Chicago, IL, 60637, USA.
| |
Collapse
|
10
|
Potenza L, Luppi M, Zimmermann C, Bandieri E. Early palliative care in haematological malignancies. BMJ Support Palliat Care 2024; 14:230-232. [PMID: 38388211 PMCID: PMC11103296 DOI: 10.1136/spcare-2024-004812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 02/05/2024] [Indexed: 02/24/2024]
Affiliation(s)
- Leonardo Potenza
- Early Palliative Care Clinic, Hematology Unit and Chair, Azienda Ospedaliera Universitaria di Modena, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Mario Luppi
- Early Palliative Care Clinic, Hematology Unit and Chair, Azienda Ospedaliera Universitaria di Modena, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Camilla Zimmermann
- Department of Supportive Care and Department of Medicine, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
| | - Elena Bandieri
- Early Palliative Care Clinic and Medical Oncology, Civil Hospital Carpi, Local Health Agency (USL), Carpi (MO), Italy
| |
Collapse
|
11
|
Niscola P, Gianfelici V, Giovannini M, Piccioni D, Mazzone C, de Fabritiis P. Latest Insights and Therapeutic Advances in Myelodysplastic Neoplasms. Cancers (Basel) 2024; 16:1563. [PMID: 38672645 PMCID: PMC11048617 DOI: 10.3390/cancers16081563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Myelodysplastic syndromes/neoplasms (MDSs) encompass a range of hematopoietic malignancies, commonly affecting elderly individuals. Molecular alterations in the hematopoietic stem cell compartment drive disease pathogenesis. Recent advancements in genomic profiling have provided valuable insights into the biological underpinnings of MDSs and have expanded therapeutic options, particularly for specific molecularly defined subgroups. This review highlights the diagnostic principles, classification updates, prognostic stratification systems, and novel treatments, which could inform future clinical trials and enhance the management of adult MDS patients, particularly for specific molecularly defined subgroups.
Collapse
Affiliation(s)
- Pasquale Niscola
- Division of Haematology, Sant’ Eugenio Hospital, 00144 Rome, Italy; (V.G.); (M.G.); (D.P.); (C.M.); (P.d.F.)
| | | | | | | | | | | |
Collapse
|
12
|
Li D, Yuan Y, Meng C, Lin Z, Zhao M, Shi L, Li M, Ye D, Cai Y, He X, Ye H, Zhou S, Zhou H, Gao S. Low expression of miR-182 caused by DNA hypermethylation accelerates acute lymphocyte leukemia development by targeting PBX3 and BCL2: miR-182 promoter methylation is a predictive marker for hypomethylation agents + BCL2 inhibitor venetoclax. Clin Epigenetics 2024; 16:48. [PMID: 38528641 PMCID: PMC10964616 DOI: 10.1186/s13148-024-01658-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 03/14/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND miR-182 promoter hypermethylation frequently occurs in various tumors, including acute myeloid leukemia, and leads to low expression of miR-182. However, whether adult acute lymphocyte leukemia (ALL) cells have high miR-182 promoter methylation has not been determined. METHODS To assess the methylation status of the miR-182 promoter, methylation and unmethylation-specific PCR analysis, bisulfite-sequencing analysis, and MethylTarget™ assays were performed to measure the frequency of methylation at the miR-182 promoter. Bone marrow cells were isolated from miR-182 knockout (182KO) and 182 wild type (182WT) mice to construct BCR-ABL (P190) and Notch-induced murine B-ALL and T-ALL models, respectively. Primary ALL samples were performed to investigate synergistic effects of the hypomethylation agents (HMAs) and the BCL2 inhibitor venetoclax (Ven) in vitro. RESULTS miR-182 (miR-182-5P) expression was substantially lower in ALL blasts than in normal controls (NCs) because of DNA hypermethylation at the miR-182 promoter in ALL blasts but not in normal controls (NCs). Knockout of miR-182 (182KO) markedly accelerated ALL development, facilitated the infiltration, and shortened the OS in a BCR-ABL (P190)-induced murine B-ALL model. Furthermore, the 182KO ALL cell population was enriched with more leukemia-initiating cells (CD43+B220+ cells, LICs) and presented higher leukemogenic activity than the 182WT ALL population. Furthermore, depletion of miR-182 reduced the OS in a Notch-induced murine T-ALL model, suggesting that miR-182 knockout accelerates ALL development. Mechanistically, overexpression of miR-182 inhibited proliferation and induced apoptosis by directly targeting PBX3 and BCL2, two well-known oncogenes, that are key targets of miR-182. Most importantly, DAC in combination with Ven had synergistic effects on ALL cells with miR-182 promoter hypermethylation, but not on ALL cells with miR-182 promoter hypomethylation. CONCLUSIONS Collectively, we identified miR-182 as a tumor suppressor gene in ALL cells and low expression of miR-182 because of hypermethylation facilitates the malignant phenotype of ALL cells. DAC + Ven cotreatment might has been applied in the clinical try for ALL patients with miR-182 promoter hypermethylation. Furthermore, the methylation frequency at the miR-182 promoter should be a potential biomarker for DAC + Ven treatment in ALL patients.
Collapse
Affiliation(s)
- Danyang Li
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China
| | - Yigang Yuan
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China
| | - Chen Meng
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China
| | - Zihan Lin
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China
| | - Min Zhao
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China
| | - Liuzhi Shi
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, Zhejiang Province, China
| | - Min Li
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China
| | - Daijiao Ye
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China
| | - Yue Cai
- Department of Clinical Medicine, Wenzhou Medical University, Chashan District, Wenzhou, Zhejiang Province, China
| | - Xiaofei He
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China
- The Key Laboratory of Pediatric Hematology and Oncology Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xuanyuanxi Road, Wenzhou, Zhejiang Province, China
| | - Haige Ye
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, Zhejiang Province, China
| | - Shujuan Zhou
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, Zhejiang Province, China
| | - Haixia Zhou
- The Key Laboratory of Pediatric Hematology and Oncology Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xuanyuanxi Road, Wenzhou, Zhejiang Province, China.
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xuanyuanxi Road, Wenzhou, Zhejiang Province, China.
| | - Shenmeng Gao
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China.
- The Key Laboratory of Pediatric Hematology and Oncology Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xuanyuanxi Road, Wenzhou, Zhejiang Province, China.
| |
Collapse
|