1
|
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
|
2
|
Soltani M, Sharifi MJ, Khalilian P, Sharifi M, Nematollahi P, Shapourian H, Ganjalikhani Hakemi M. Potential Diagnostic Value of Abnormal Pyroptosis Genes Expression in Myelodysplastic Syndromes (MDS): A Primary Observational Cohort Study. Int J Hematol Oncol Stem Cell Res 2024; 18:156-164. [PMID: 38868810 PMCID: PMC11166493 DOI: 10.18502/ijhoscr.v18i2.15371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 08/06/2023] [Indexed: 06/14/2024] Open
Abstract
Background: Myelodysplastic syndromes (MDS) are determined by ineffective hematopoiesis and bone marrow cytological dysplasia with somatic gene mutations and chromosomal abnormalities. Accumulating evidence has revealed the pivotal role of NLRP3 inflammasome activation and pyroptotic cell death in the pathogenesis of MDS. Although MDS can be diagnosed with a variety of morphologic and cytogenetic tests, most of these tests have limitations or problems in practice. Materials and Methods: In the present study, we evaluated the expression of genes that form the inflammasome (NLRP3, ASC, and CASP1) in bone marrow specimens of MDS patients and compared the results with those of other leukemias to evaluate their diagnostic value for MDS. Primary samples of this observational cohort study were collected from aspiration samples of patients with myelodysplastic syndromes (27 cases) and patients with non-myelodysplastic syndrome hematological cancers (45 cases). After RNA extraction and c.DNA synthesis, candidate transcripts and housekeeping transcripts were measured by real-time PCR method (SYBER Green assay). Using Kruskal-Wallis the relative gene expressions were compared and differences with p value less than 0.05 were considered as significant. Discrimination capability, cut-off, and area under curve (AUC) of all markers were analyzed with recessive operation curve (ROC) analysis. Results: We found that Caspase-1 and ASC genes expressed at more levels in MDS specimens compared to non-MDS hematological malignancies. A relative average expression of 10.22 with a p-value of 0.001 and 1.86 with p=0.019 was detected for Caspase-1 and ASC, respectively. ROC curve analysis shows an AUC of 0.739 with p=0.0001 for Caspase-1 and an AUC of 0.665 with p=0.0139 for ASC to MDS discrimination. Conclusion: Our results show that Caspase-1 and ASC gene expression levels can be used as potential biomarkers for MDS diagnosis. Prospective studies with large sample numbers are suggested.
Collapse
Affiliation(s)
- Mohammad Soltani
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Jafar Sharifi
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parvin Khalilian
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehran Sharifi
- Department of Internal Medicine, School of Medicine, cancer Prevention Research Center, Seyyed Al-Shohada Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Pardis Nematollahi
- Department of Pathology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hooriyeh Shapourian
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mazdak Ganjalikhani Hakemi
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute of Health sciences and Technology (SABITA), Istanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
3
|
Maurya N, Shanmukhaiah C, Dhangar S, Madkaikar M, Vundinti BR. Comprehensive Study of Chromosomal Copy Number Variations and Genomic Variations Predicting Overall Survival in Myelodysplastic Syndromes. Oncology 2024; 102:897-906. [PMID: 38442690 DOI: 10.1159/000536446] [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: 11/07/2023] [Accepted: 01/13/2024] [Indexed: 03/07/2024]
Abstract
INTRODUCTION Myelodysplastic syndrome (MDS) is a heterogeneous disease characterized by cytopenia, marrow dysplasia and has a propensity to develop into acute myeloid leukemia. The disease progression is majorly affected by genetic defects. However, about 40-50% of patients with MDS present with a normal karyotype and develop different courses of disease. Hence, there remains a room to advance the biological understanding and find molecular prognostic markers for cytogenetically normal MDS. METHODS We performed a high-resolution CGH + SNP array along with next-generation sequencing (NGS) of 77 primary diagnosed MDS patients, and also they were clinically followed up. RESULTS Our study revealed 82 clinically significant genomic lesions (losses/gains) in 49% of MDS patients. CGH + SNP array reduced the proportion of normal karyotype by 30%. SNP array in combination with NGS confirmed the biallelic loss of function of the TP53 gene (2/6), which is a clinically relevant biomarker and new genetic-based MDS entity, i.e., MDS-biTP53, as per the new WHO classification 2022. Genomic region 2p22.3 presented with frequent lesions and also with a more hazard ratio (2.7, 95% CI: 0.37-21) when analyzed by Kaplan-Meier survival analysis. CONCLUSION CGH + SNP array changed the cytogenetic and IPSS-R risk group in 18% and 13% of patients, respectively, with an improved prediction of prognosis. This study emphasizes the cytogenetic heterogeneity of MDS and highlights that abnormality with chromosome 2 may have a diagnostic and prognostic impact.
Collapse
Affiliation(s)
- Nehakumari Maurya
- Department of Cytogenetics, ICMR-National Institute of Immunohaematology, Mumbai, India
| | | | - Somprakash Dhangar
- Department of Cytogenetics, ICMR-National Institute of Immunohaematology, Mumbai, India
| | - Manisha Madkaikar
- Department of Cytogenetics, ICMR-National Institute of Immunohaematology, Mumbai, India
| | - Babu Rao Vundinti
- Department of Cytogenetics, ICMR-National Institute of Immunohaematology, Mumbai, India
| |
Collapse
|
4
|
Sinha R, Dvorak M, Ganesan A, Kalesinskas L, Niemeyer CM, Flotho C, Sakamoto KM, Lacayo N, Patil RV, Perriman R, Cepika AM, Liu YL, Kuo A, Utz PJ, Khatri P, Bertaina A. Epigenetic Profiling of PTPN11 Mutant JMML Hematopoietic Stem and Progenitor Cells Reveals an Aberrant Histone Landscape. Cancers (Basel) 2023; 15:5204. [PMID: 37958378 PMCID: PMC10650722 DOI: 10.3390/cancers15215204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 10/18/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Juvenile myelomonocytic leukemia (JMML) is a deadly pediatric leukemia driven by RAS pathway mutations, of which >35% are gain-of-function in PTPN11. Although DNA hypermethylation portends severe clinical phenotypes, the landscape of histone modifications and chromatin profiles in JMML patient cells have not been explored. Using global mass cytometry, Epigenetic Time of Flight (EpiTOF), we analyzed hematopoietic stem and progenitor cells (HSPCs) from five JMML patients with PTPN11 mutations. These data revealed statistically significant changes in histone methylation, phosphorylation, and acetylation marks that were unique to JMML HSPCs when compared with healthy controls. Consistent with these data, assay for transposase-accessible chromatin with sequencing (ATAC-seq) analysis revealed significant alterations in chromatin profiles at loci encoding post-translational modification enzymes, strongly suggesting their mis-regulated expression. Collectively, this study reveals histone modification pathways as an additional epigenetic abnormality in JMML patient HSPCs, thereby uncovering a new family of potential druggable targets for the treatment of JMML.
Collapse
Affiliation(s)
- Roshani Sinha
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA 94305, USA; (R.S.); (R.V.P.); (R.P.); (A.-M.C.); (Y.L.L.)
| | - Mai Dvorak
- Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (M.D.); (A.G.); (L.K.); (A.K.); (P.J.U.); (P.K.)
| | - Ananthakrishnan Ganesan
- Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (M.D.); (A.G.); (L.K.); (A.K.); (P.J.U.); (P.K.)
| | - Larry Kalesinskas
- Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (M.D.); (A.G.); (L.K.); (A.K.); (P.J.U.); (P.K.)
| | - Charlotte M. Niemeyer
- Department of Pediatric Hematology and Oncology, University of Freiburg Medical Centre, 79098 Freiburg im Breisgau, Germany; (C.M.N.); (C.F.)
| | - Christian Flotho
- Department of Pediatric Hematology and Oncology, University of Freiburg Medical Centre, 79098 Freiburg im Breisgau, Germany; (C.M.N.); (C.F.)
| | - Kathleen M. Sakamoto
- Bass Center for Childhood Cancer and Blood Disorders at Lucile Packard Children’s Hospital, Palo Alto, CA 94304, USA; (K.M.S.); (N.L.)
| | - Norman Lacayo
- Bass Center for Childhood Cancer and Blood Disorders at Lucile Packard Children’s Hospital, Palo Alto, CA 94304, USA; (K.M.S.); (N.L.)
| | - Rachana Vinay Patil
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA 94305, USA; (R.S.); (R.V.P.); (R.P.); (A.-M.C.); (Y.L.L.)
| | - Rhonda Perriman
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA 94305, USA; (R.S.); (R.V.P.); (R.P.); (A.-M.C.); (Y.L.L.)
| | - Alma-Martina Cepika
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA 94305, USA; (R.S.); (R.V.P.); (R.P.); (A.-M.C.); (Y.L.L.)
| | - Yunying Lucy Liu
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA 94305, USA; (R.S.); (R.V.P.); (R.P.); (A.-M.C.); (Y.L.L.)
| | - Alex Kuo
- Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (M.D.); (A.G.); (L.K.); (A.K.); (P.J.U.); (P.K.)
| | - Paul J. Utz
- Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (M.D.); (A.G.); (L.K.); (A.K.); (P.J.U.); (P.K.)
| | - Purvesh Khatri
- Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (M.D.); (A.G.); (L.K.); (A.K.); (P.J.U.); (P.K.)
| | - Alice Bertaina
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA 94305, USA; (R.S.); (R.V.P.); (R.P.); (A.-M.C.); (Y.L.L.)
- Bass Center for Childhood Cancer and Blood Disorders at Lucile Packard Children’s Hospital, Palo Alto, CA 94304, USA; (K.M.S.); (N.L.)
| |
Collapse
|
5
|
Kontandreopoulou CN, Kalopisis K, Viniou NA, Diamantopoulos P. The genetics of myelodysplastic syndromes and the opportunities for tailored treatments. Front Oncol 2022; 12:989483. [PMID: 36338673 PMCID: PMC9630842 DOI: 10.3389/fonc.2022.989483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Genomic instability, microenvironmental aberrations, and somatic mutations contribute to the phenotype of myelodysplastic syndrome and the risk for transformation to AML. Genes involved in RNA splicing, DNA methylation, histone modification, the cohesin complex, transcription, DNA damage response pathway, signal transduction and other pathways constitute recurrent mutational targets in MDS. RNA-splicing and DNA methylation mutations seem to occur early and are reported as driver mutations in over 50% of MDS patients. The improved understanding of the molecular landscape of MDS has led to better disease and risk classification, leading to novel therapeutic opportunities. Based on these findings, novel agents are currently under preclinical and clinical development and expected to improve the clinical outcome of patients with MDS in the upcoming years. This review provides a comprehensive update of the normal gene function as well as the impact of mutations in the pathogenesis, deregulation, diagnosis, and prognosis of MDS, focuses on the most recent advances of the genetic basis of myelodysplastic syndromes and their clinical relevance, and the latest targeted therapeutic approaches including investigational and approved agents for MDS.
Collapse
|
6
|
Maioli M, Rinaldi S, Cruciani S, Necas A, Fontani V, Corda G, Santaniello S, Rinaldi A, Barcessat A, Necasova A, Castagna A, Filipejova Z, Ventura C, Fozza C. Antisenescence effect of REAC biomodulation to counteract the evolution of myelodysplastic syndrome. Physiol Res 2022; 71:539-549. [PMID: 35899943 PMCID: PMC9616590 DOI: 10.33549/physiolres.934903] [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: 03/18/2022] [Accepted: 07/15/2022] [Indexed: 11/25/2022] Open
Abstract
About 30 percent of patients diagnosed with myelodysplastic syndromes (MDS) progress to acute myeloid leukemia (AML). The senescence of bone marrow?derived mesenchymal stem cells (BMSCs) seems to be one of the determining factors in inducing this drift. Research is continuously looking for new methodologies and technologies that can use bioelectric signals to act on senescence and cell differentiation towards the phenotype of interest. The Radio Electric Asymmetric Conveyer (REAC) technology, aimed at reorganizing the endogenous bioelectric activity, has already shown to be able to determine direct cell reprogramming effects and counteract the senescence mechanisms in stem cells. Aim of the present study was to prove if the anti-senescence results previously obtained in different kind of stem cells with the REAC Tissue optimization - regenerative (TO-RGN) treatment, could also be observed in BMSCs, evaluating cell viability, telomerase activity, p19ARF, P21, P53, and hTERT gene expression. The results show that the REAC TO-RGN treatment may be a useful tool to counteract the BMSCs senescence which can be the basis of AML drift. Nevertheless, further clinical studies on humans are needed to confirm this hypothesis.
Collapse
Affiliation(s)
- M Maioli
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Zamora DI, Patel GS, Grossmann I, Rodriguez K, Soni M, Joshi PK, Patel SC, Shreya D, Sange I. Myelodysplastic Syndromes and Modalities of Treatment: An Updated Literature Review. Cureus 2021; 13:e20116. [PMID: 34873563 PMCID: PMC8639322 DOI: 10.7759/cureus.20116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2021] [Indexed: 11/29/2022] Open
Abstract
Myelodysplastic syndromes (MDS) represent a large group of rare and diverse clonal stem cell disorders. These are classified into several different phenotypes and typically arise following a multistep genetic process, whereby genetic mutations alter the DNA damage and cellular stress responses, impacting transcription, RNA splicing, epigenetics, and cytokine signaling. However, despite the advances made regarding molecular pathophysiology and prognostic criteria and the influx of new treatment modalities, management is primarily based on prognostic scores, such as the Revised International Prognostic Scoring System. This poses a significant challenge to current healthcare professionals due to poor comprehension of the underlying pathophysiology. Hence, this review integrates the latest research and treatment modalities for MDS and discusses the different genetic mutations outlined in the revised World Health Organization 2016 MDS classification system and the associated treatment modalities. Additionally, future directions of research and clinical management of MDS are discussed.
Collapse
Affiliation(s)
- Diana I Zamora
- General Medicine, Universidad de Ciencias Médicas, San José, CRI
| | - Gautami S Patel
- Internal Medicine, Pramukhswami Medical College, Karamsad, IND
| | - Idan Grossmann
- Research, Medical University of Silesia, Faculty of Medical Sciences in Katowice, Katowice, POL
| | - Kevin Rodriguez
- Research, Universidad Americana Facultad de Medicina, Managua, NIC
| | - Mridul Soni
- Research, Shri Lal Bahadur Shastri Government Medical College, Mandi, IND
| | - Pranay K Joshi
- Department of Medicine, B.J. Medical College, Ahmedabad, IND
| | | | | | | |
Collapse
|
8
|
Lee P, Yim R, Yung Y, Chu HT, Yip PK, Gill H. Molecular Targeted Therapy and Immunotherapy for Myelodysplastic Syndrome. Int J Mol Sci 2021; 22:10232. [PMID: 34638574 PMCID: PMC8508686 DOI: 10.3390/ijms221910232] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 12/22/2022] Open
Abstract
Myelodysplastic syndrome (MDS) is a heterogeneous, clonal hematological disorder characterized by ineffective hematopoiesis, cytopenia, morphologic dysplasia, and predisposition to acute myeloid leukemia (AML). Stem cell genomic instability, microenvironmental aberrations, and somatic mutations contribute to leukemic transformation. The hypomethylating agents (HMAs), azacitidine and decitabine are the standard of care for patients with higher-risk MDS. Although these agents induce responses in up to 40-60% of patients, primary or secondary drug resistance is relatively common. To improve the treatment outcome, combinational therapies comprising HMA with targeted therapy or immunotherapy are being evaluated and are under continuous development. This review provides a comprehensive update of the molecular pathogenesis and immune-dysregulations involved in MDS, mechanisms of resistance to HMA, and strategies to overcome HMA resistance.
Collapse
Affiliation(s)
| | | | | | | | | | - Harinder Gill
- Division of Haematology, Medical Oncology and Haemopoietic Stem Cell Transplantation, Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (P.L.); (R.Y.); (Y.Y.); (H.-T.C.); (P.-K.Y.)
| |
Collapse
|
9
|
EnvIRONmental Aspects in Myelodysplastic Syndrome. Int J Mol Sci 2021; 22:ijms22105202. [PMID: 34068996 PMCID: PMC8156755 DOI: 10.3390/ijms22105202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 11/24/2022] Open
Abstract
Systemic iron overload is multifactorial in patients suffering from myelodysplastic syndrome (MDS). Disease-immanent ineffective erythropoiesis together with chronic red blood cell transfusion represent the main underlying reasons. However, like the genetic heterogeneity of MDS, iron homeostasis is also diverse in different MDS subtypes and can no longer be generalized. While a certain amount of iron and reactive oxygen species (ROS) are indispensable for proper hematological output, both are harmful if present in excess. Consequently, iron overload has been increasingly recognized as an important player in MDS, which is worth paying attention to. This review focuses on iron- and ROS-mediated effects in the bone marrow niche, their implications for hematopoiesis and their yet unclear involvement in clonal evolution. Moreover, we provide recent insights into hepcidin regulation in MDS and its interaction between erythropoiesis and inflammation. Based on Tet methylcytosine dioxygenase 2 (TET2), representing one of the most frequently mutated genes in MDS, leading to disturbances in both iron homeostasis and hematopoiesis, we highlight that different genetic alteration may have different implications and that a comprehensive workup is needed for a complete understanding and development of future therapies.
Collapse
|
10
|
Zhu H, Yang B, Liu J, Wang B, Wu Y, Zheng Z, Ling Y. A novel treatment regimen of granulocyte colony-stimulating factor combined with ultra-low-dose decitabine and low-dose cytarabine in older patients with acute myeloid leukemia and myelodysplastic syndromes. Ther Adv Hematol 2021; 12:20406207211009334. [PMID: 33995987 PMCID: PMC8111530 DOI: 10.1177/20406207211009334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 03/21/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Older patients with acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) unfit for intensive chemotherapy are emergent for suitable treatment strategies. Hypomethylating agents and low-dose cytarabine have generated relevant benefits in the hematological malignancies over recent decades. We evaluated the efficacy and safety of the novel treatment regimen consisting of ultra-low-dose decitabine and low-dose cytarabine, with granulocyte colony-stimulating factor (G-CSF) in this population of patients. Methods and materials: Patients aged more than 60 years with newly diagnosed AML/MDS were enrolled to receive therapy combined of 300 µg subcutaneously per day for priming, decitabine 5.15–7.62 mg/m2/d intravenously and cytarabine 15 mg/m2/d twice a day subcutaneously and G-CSF for consecutive 10 days every 28 days. The study enrolled 28 patients unfit for standard intensive chemotherapy. The median age of patients was 68 years (range 60–83 years) and 20 (71.4%) patients harbored AML. The primary outcome was to evaluate overall response rate. Results: Overall, this novel ultra-low-dose treatment regimen was well tolerated, with 0% of both 4- and 8-week mortality occurrence. Objective response rate (CR + CRi + PR in AML and CR + mCR + PR in MDS) was 57.1% after the first treatment course. Responses of hematologic improvement (HI) aspect were achieved in 18 of 28 (64.3%) patients, 11 (39.3%), 12 (42.9%), and eight patients (28.6%) achieved HI-E, HI-P, HI-N, respectively. Conclusions: Untreated elderly with AML/MDS were well tolerated and benefited from this novel ultra-low-dose treatment regimen.
Collapse
Affiliation(s)
- Huan Zhu
- Department of Hematology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Bin Yang
- The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Jia Liu
- The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Biao Wang
- The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Yicun Wu
- The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Zhuojun Zheng
- The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Yun Ling
- Department of Hematology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, Juqian Road 185, Changzhou, Jiangsu 213000, China
| |
Collapse
|
11
|
Current State and Challenges in Development of Targeted Therapies in Myelodysplastic Syndromes (MDS). HEMATO 2021. [DOI: 10.3390/hemato2020013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Myelodysplastic syndromes (MDS) encompass a variety of myeloid neoplasms characterized by ineffective hematopoiesis. The interaction of abnormal clonal hematopoiesis and changes in the bone marrow microenvironment propagate abnormal clones. Advances in next generation sequencing has identified over 100 somatic mutations, but despite deepened understanding of the genetics of MDS, therapeutic discoveries have remained limited. To date, only five drugs have been approved for MDS: Azacitidine, Decitabine, Lenalidomide, Luspatercept, and oral Decitabine with Cedazuridine. Current strategies for low-risk MDS continue to focus on symptomatic management and correction of cytopenias, while treatment for high-risk MDS focuses on delaying progression of disease and improving survival. In this review we discuss some of the challenges in developing pre-clinical models of MDS in which to test therapeutics, the advances that have been made, and promising novel therapeutics in the pipeline.
Collapse
|
12
|
Sundaravel S, Steidl U, Wickrema A. Epigenetic modifiers in normal and aberrent erythropoeisis. Semin Hematol 2021; 58:15-26. [PMID: 33509439 PMCID: PMC7883935 DOI: 10.1053/j.seminhematol.2020.12.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022]
Abstract
Erythroid differentiation program is comprised of lineage commitment, erythroid progenitor proliferation, and termination differentiation. Each stage of the differentiation program is heavily influenced by epigenetic modifiers that alter the epigenome in a dynamic fashion influenced by cytokines/humeral factors and are amicable to target by drugs. The epigenetic modifiers can be classified as DNA modifiers (DNMT, TET), mRNA modifiers (RNA methylases and demethylases) and histone protein modifiers (methyltransferases, acetyltransferases, demethylases, and deacetylases). Here we describe mechanisms by which these epigenetic modifiers influence and guide erythroid-lineage differentiation during normal and malignant erythropoiesis and also benign diseases that arise from their altered structure or function.
Collapse
Affiliation(s)
- Sriram Sundaravel
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Ulrich Steidl
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY; Department of Medicine, Albert Einstein College of Medicine-Montefiore Medical center, Bronx, NY
| | | |
Collapse
|
13
|
Cavalcante GM, Borges DP, de Oliveira RTG, Furtado CLM, Alves APNN, Sousa AM, de Paula DS, Filho FDR, Magalhães SMM, Ribeiro-Jr HL, Pinheiro RF. Tissue methylation and demethylation influence translesion synthesis DNA polymerases (TLS) contributing to the genesis of chromosomal abnormalities in myelodysplastic syndrome. J Clin Pathol 2020; 75:85-93. [PMID: 33234697 DOI: 10.1136/jclinpath-2020-207131] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/21/2020] [Accepted: 11/05/2020] [Indexed: 12/19/2022]
Abstract
AIMS DNA methylation has its distribution influenced by DNA demethylation processes with the catalytic conversion of 5-methylcytosine (5mC) into 5-hydroxymethylcytosine (5hmC). Myelodysplastic syndrome (MDS) has been associated with epigenetic dysregulation of genes related to DNA repair system, chronic immune response and cell cycle. METHODS We evaluated the tissue DNA methylation/hydroxymethylation in bone marrow trephine biopsies of 73 patients with MDS, trying to correlate with the mRNA expression of 21 genes (POLH, POLL, REV3L, POLN, POLQ, POLI, POLK, IRF-1, IRF-2, IRF-3, IRF-4, IRF-5, IRF6, IRF-7, IRF-8,IRF-9, MAD2, CDC20, AURKA, AURKB and TPX2). RESULTS The M-score (5mC) was significantly higher in patients with chromosomal abnormalities than patients with normal karyotype (95% CI -27.127779 to -2.368020; p=0.022). We observed a higher 5mC/5hmC ratio in patients classified as high-risk subtypes compared with low-risk subtypes (95% CI -72.922115 to -1.855662; p=0.040) as well as patients with hypercellular bone marrow compared with patients with normocellular/hypocellular bone marrow (95% CI -69.189259 to -0.511828; p=0.047) and with the presence of dyserythropoiesis (95% CI 17.077703 to 51.331388; p=0.001). DNA pols with translesion activity are significantly influenced by methylation. As 5mC immunoexpression increases, the expressions of POLH (r=-0.816; r2 =0.665; p=0.000), POLQ (r=-0.790; r2=0.624; p=0.001), PCNA (r=-0.635; r2=0.403; p=0.020), POLK (r=-0.633; r2=0.400; p=0.036 and REV1 (r=-0.578; r2=0.334; p=0.049) decrease. CONCLUSIONS Our results confirm that there is an imbalance in the DNA methylation in MDS, influencing the development of chromosomal abnormalities which may be associated with the low expression of DNA polymerases with translesion synthesis polymerases activity.
Collapse
Affiliation(s)
- Gabrielle Melo Cavalcante
- Postgraduate Program in Pathology, Federal University of Ceara, Fortaleza, Ceara, Brazil.,Cancer Cytogenomic Laboratory, Drug Research and Development Center, Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Daniela Paula Borges
- Cancer Cytogenomic Laboratory, Drug Research and Development Center, Federal University of Ceara, Fortaleza, Ceara, Brazil.,Postgraduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Roberta Taiane Germano de Oliveira
- Cancer Cytogenomic Laboratory, Drug Research and Development Center, Federal University of Ceara, Fortaleza, Ceara, Brazil.,Postgraduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Cristiana Libardi Miranda Furtado
- Postgraduate Program in Medical and Surgical Sciences, Universidade Federal do Ceara, Fortaleza, Ceara, Brazil.,Drug Research and Development Center, Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Ana Paula Negreiros Nunes Alves
- Department of Dental Clinic, Faculty of Pharmacy, Dentistry and Nursing, Universidade Federal do Ceara, Fortaleza, Ceara, Brazil
| | - Alceu Machado Sousa
- Department of Odontology Clinic, Universidade Federal do Ceara, Fortaleza, Ceara, Brazil
| | | | | | - Silvia Maria Meira Magalhães
- Cancer Cytogenomic Laboratory, Drug Research and Development Center, Federal University of Ceara, Fortaleza, Ceara, Brazil.,Postgraduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Howard Lopes Ribeiro-Jr
- Postgraduate Program in Pathology, Federal University of Ceara, Fortaleza, Ceara, Brazil.,Cancer Cytogenomic Laboratory, Drug Research and Development Center, Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Ronald Feitosa Pinheiro
- Postgraduate Program in Pathology, Federal University of Ceara, Fortaleza, Ceara, Brazil .,Cancer Cytogenomic Laboratory, Drug Research and Development Center, Federal University of Ceara, Fortaleza, Ceara, Brazil.,Postgraduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil
| |
Collapse
|
14
|
Lee DY. Cancer Epigenomics and Beyond: Advancing the Precision Oncology Paradigm. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2020; 3:147-156. [PMID: 35665374 PMCID: PMC9165444 DOI: 10.36401/jipo-20-18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 07/14/2020] [Indexed: 06/15/2023]
Abstract
How cancers are characterized and treated has evolved over the past few decades. Major advances in genomics tools and techniques have revealed interlinked regulatory pathways of cancers with unprecedented detail. Early discoveries led to success with rationally targeted small molecules and more recently with immunomodulatory agents, setting the stage for precision oncology. However, drug resistance to every agent has thus far proven intractable, sending us back to fill the gaps in our rudimentary knowledge of tumor biology. Epigenetics is emerging as a fundamental process in every hallmark of cancer. Large-scale interrogation of the cancer epigenome continues to reveal new mechanisms of astounding complexity. In this review, I present selected experimental and clinical examples that have shaped our understanding of cancer at the molecular level. Translation of our collective erudition into revolutionary diagnostic and treatment strategies will advance the precision oncology paradigm.
Collapse
Affiliation(s)
- Daniel Y. Lee
- InSilico Genomics, Inc., Houston, TX, USA
- Department of Radiology, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
15
|
Derissen EJB, Beijnen JH. Intracellular Pharmacokinetics of Pyrimidine Analogues used in Oncology and the Correlation with Drug Action. Clin Pharmacokinet 2020; 59:1521-1550. [PMID: 33064276 PMCID: PMC7717039 DOI: 10.1007/s40262-020-00934-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pyrimidine analogues can be considered as prodrugs, like their natural counterparts, they have to be activated within the cell. The intracellular activation involves several metabolic steps including sequential phosphorylation to its monophosphate, diphosphate and triphosphate. The intracellularly formed nucleotides are responsible for the pharmacological effects. This review provides a comprehensive overview of the clinical studies that measured the intracellular nucleotide concentrations of pyrimidine analogues in patients with cancer. The objective was to gain more insight into the parallels between the different pyrimidine analogues considering their intracellular pharmacokinetics. For cytarabine and gemcitabine, the intracellular pharmacokinetics have been extensively studied over the years. However, for 5-fluorouracil, capecitabine, azacitidine and decitabine, the intracellular pharmacokinetics was only very minimally investigated. This is probably owing to the fact that there were no suitable bioanalytical assays for a long time. Since the advent of suitable assays, the first exploratory studies indicate that the intracellular 5-fluorouracil, azacitidine and decitabine nucleotide concentrations are very low compared with the intracellular nucleotide concentrations obtained during treatment with cytarabine or gemcitabine. Based on their pharmacology, the intracellular accumulation of nucleotides appears critical to the cytotoxicity of pyrimidine analogues. However, not many clinical studies have actually investigated the relationship between the intracellular nucleotide concentrations in patients with cancer and the anti-tumour effect. Only for cytarabine, a relationship was demonstrated between the intracellular triphosphate concentrations in leukaemic cells and the response rate in patients with AML. Future clinical studies should show, for the other pyrimidine analogues, whether there is a relationship between the intracellular nucleotide concentrations and the clinical outcome of patients. Research that examined the intracellular pharmacokinetics of cytarabine and gemcitabine focused primarily on the saturation aspect of the intracellular triphosphate formation. Attempts to improve the dosing regimen of gemcitabine were aimed at maximising the intracellular gemcitabine triphosphate concentrations. However, this strategy does not make sense, as efficient administration also means that less gemcitabine can be administered before dose-limiting toxicities are achieved. For all pyrimidine analogues, a linear relationship was found between the dose and the plasma concentration. However, no correlation was found between the plasma concentration and the intracellular nucleotide concentration. The concentration-time curves for the intracellular nucleotides showed considerable inter-individual variation. Therefore, the question arises whether pyrimidine analogue therapy should be more individualised. Future research should show which intracellular nucleotide concentrations are worth pursuing and whether dose individualisation is useful to achieve these concentrations.
Collapse
Affiliation(s)
- Ellen J B Derissen
- Department of Pharmacy and Pharmacology, Antoni van Leeuwenhoek Hospital-The Netherlands Cancer Institute, Louwesweg 6, 1066 EC , Amsterdam, The Netherlands. .,Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands. .,Department of Pharmacy , Elisabeth-TweeSteden Hospital, Dr. Deelenlaan 5, 5042 AD, Tilburg, The Netherlands.
| | - Jos H Beijnen
- Department of Pharmacy and Pharmacology, Antoni van Leeuwenhoek Hospital-The Netherlands Cancer Institute, Louwesweg 6, 1066 EC , Amsterdam, The Netherlands.,Science Faculty, Division of Pharmaco-epidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, P.O. Box 80082, 3508 TB, Utrecht, The Netherlands
| |
Collapse
|
16
|
Li H, Wang Y, Pang X, Xie C, Deeg HJ, Wang H, Wan T, Wu J, Guan F, Li X. Elevated TWIST1 expression in myelodysplastic syndromes/acute myeloid leukemia reduces efficacy of hypomethylating therapy with decitabine. Haematologica 2020; 105:e502. [PMID: 33054092 PMCID: PMC7556660 DOI: 10.3324/haematol.2019.235325] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Affiliation(s)
- Hongjiao Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, China
| | - Yi Wang
- Department of Hematology, Provincial People’s Hospital, Xi’an, Shaanxi, China
| | - Xingchen Pang
- School of Biotechnology, Jiangnan University, Wuxi, China
| | - Chenglian Xie
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, China
| | - H. Joachim Deeg
- Fred Hutchinson Cancer Research Center, and University of Washington School of Medicine, Seattle, WA, USA
| | - Hui Wang
- Department of Hematology, Provincial People’s Hospital, Xi’an, Shaanxi, China
| | - Ting Wan
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jinpeng Wu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, China
| | - Feng Guan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, China
| | - Xiang Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, China
| |
Collapse
|
17
|
IGF‑IR promotes clonal cell proliferation in myelodysplastic syndromes via inhibition of the MAPK pathway. Oncol Rep 2020; 44:1094-1104. [PMID: 32583001 PMCID: PMC7388562 DOI: 10.3892/or.2020.7652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 05/19/2020] [Indexed: 12/12/2022] Open
Abstract
Type 1 insulin-like growth factor receptor (IGF-IR) signaling is considered to serve a key role in the development of cancer. However, the effects of IGF-IR on the malignant characteristics of myelodysplastic syndrome (MDS) clonal cells remains to be determined. In the present study it was demonstrated that knockdown of IGF-IR reduced the proliferation and increased the apoptosis of MDS/leukemia cells. Integrated analysis of gene expression profiles using bioinformatics identified the MAPK signaling pathway as a critical downstream factor of IGF-IR, and this was confirmed in vitro using western blotting which revealed that IGF-IR knockdown significantly increased the expression of activated MAPK. Furthermore, IGF-IR signaling was inhibited to investigate the potential of IGF-IR as a therapeutic target of MDS. The results revealed that the IGF-IR inhibitor picropodophyllin (PPP) inhibited cell proliferation, promoted cell apoptosis and arrested the cell cycle at the G2/M phase in MDS/leukemia cells. Similar to the effects of IGF-IR knockdown, PPP treatment also increased MAPK signaling in vitro. In conclusion, IGF-IR may serve as a potential therapeutic target of MDS.
Collapse
|
18
|
Badar T, Szabo A, Sallman D, Komrojki R, Lancet J, Padron E, Song J, Hussaini MO. Interrogation of molecular profiles can help in differentiating between MDS and AML with MDS-related changes. Leuk Lymphoma 2020; 61:1418-1427. [DOI: 10.1080/10428194.2020.1719089] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Talha Badar
- Division of Hematology and Oncology, Medical College of Wisconsin and Froedtert Hospital, Milwaukee, WI, USA
| | - Aniko Szabo
- Division of Hematology and Oncology, Medical College of Wisconsin and Froedtert Hospital, Milwaukee, WI, USA
| | - David Sallman
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| | - Rami Komrojki
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| | - Jefferey Lancet
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| | - Eric Padron
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| | - Jinming Song
- Department of Hematopathology and Laboratory Medicine, Moffitt Cancer Center, Tampa, FL, USA
| | - Mohammad Omar Hussaini
- Department of Hematopathology and Laboratory Medicine, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
19
|
Immune Dysregulation and Recurring Mutations in Myelodysplastic Syndromes Pathogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1326:1-10. [PMID: 33385175 DOI: 10.1007/5584_2020_608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Myelodysplastic syndromes (MDS) are clonal stem cell malignancies characterized by ineffective hematopoiesis leading to peripheral cytopenias and variable risk of progression to acute myeloid leukemia. Inflammation is associated with MDS pathogenesis. Several cytokines, reactive species of oxygen/nitrogen and growth factors are directly or indirectly involved in dysfunction of the MDS bone marrow (BM) microenvironment. Mutations in genes mainly regulating RNA splicing, DNA methylation and chromatin accessibility, transcription factors, signal transduction and the response to DNA damage contribute to ineffective hematopoiesis, genomic instability and MDS development. The inflammation-associated DNA damage in hematopoietic stem cells may also contribute to MDS development and progression with aggressive clinical characteristics. Many studies have aimed at clarifying mechanisms involved in the activity of immature myeloid cells as powerful modulators of the immune response and their correlation with aging, autoimmunity, and development of cancer. In this review, we explore recent advances and accumulating evidence uniting immune dysregulation, inflammaging and recurring mutations in the pathogenesis of MDS.
Collapse
|
20
|
Garcia-Manero G, Roboz G, Walsh K, Kantarjian H, Ritchie E, Kropf P, O'Connell C, Tibes R, Lunin S, Rosenblat T, Yee K, Stock W, Griffiths E, Mace J, Podoltsev N, Berdeja J, Jabbour E, Issa JPJ, Hao Y, Keer HN, Azab M, Savona MR. Guadecitabine (SGI-110) in patients with intermediate or high-risk myelodysplastic syndromes: phase 2 results from a multicentre, open-label, randomised, phase 1/2 trial. LANCET HAEMATOLOGY 2019; 6:e317-e327. [PMID: 31060979 DOI: 10.1016/s2352-3026(19)30029-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/13/2019] [Accepted: 02/13/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Guadecitabine is a next-generation hypomethylating agent whose active metabolite decitabine has a longer in-vivo exposure time than intravenous decitabine. More effective hypomethylating agents are needed for the treatment of myelodysplastic syndromes. In the present study, we aimed to compare the activity and safety of two doses of guadecitabine in hypomethylating agent treatment-naive or relapsed or refractory patients with intermediate-risk or high-risk myelodysplastic syndromes. METHODS This phase 2 part of the phase 1/2, randomised, open-label study enrolled patients aged 18 years or older from 14 North American medical centres with International Prognostic Scoring System intermediate-1-risk, intermediate-2-risk, or high-risk myelodysplastic syndromes, or chronic myelomonocytic leukaemia. They were either hypomethylating agent treatment-naive or had relapsed or refractory disease after previous hypomethylating agent treatment as determined by the investigators' judgment. Eligible patients had Eastern Cooperative Oncology Group performance status of 0-2. Patients were randomly assigned (1:1) using a computer algorithm for dynamic randomisation to subcutaneous guadecitabine 60 or 90 mg/m2 on days 1-5 of a 28-day treatment cycle. Treatment was stratified by previous treatment with hypomethylating agents and neither patients nor investigators were masked. The primary endpoint was overall response (a composite of complete response, partial response, marrow complete response, and haematological improvement) assessed in all patients who received at least one dose of study drug. This study is registered with ClinicalTrials.gov, number NCT01261312. FINDINGS Between July 9, 2012, and April 7, 2014, 105 patients were enrolled: 55 (52%) were allocated to guadecitabine 60 mg/m2 (28 patients were treatment-naive and 27 had relapsed or refractory disease after previous hypomethylating agent treatment) and 50 (48%) patients to 90 mg/m2 (23 patients were treatment-naive and 27 had relapsed or refractory disease). Three (3%) patients of 105 did not receive study treatment and were excluded from analyses. Median follow-up was 3·2 years (IQR 2·8-3·5). The proportion of patients achieving an overall response did not significantly differ between dose groups (21 of 53 [40%, 95% CI 27-54] with 60 mg/m2 and 27 of 49 [55%, 95% CI 40-69] with 90 mg/m2; p=0·16). 25 of 49 (51%, 95% CI 36-66) patients who were treatment-naive and 23 of 53 (43%, 30-58) patients with relapsed or refractory disease achieved an overall response. The most common grade 3 or worse adverse events in both groups, regardless of relationship to treatment, were thrombocytopenia (22 [41%] of 53 patients in the 60 mg/m2 group and 28 [57%] of 49 in the 90 mg/m2 group), neutropaenia (21 [40%] and 25 [51%]), anaemia (25 [47%] and 24 [49%]), febrile neutropaenia (17 [32%] and 21 [43%]), and pneumonia (13 [25%] and 15 [31%]). Seven (7%) of 102 patients died due to adverse events (three with 90 mg/m2 and four with 60 mg/m2), and all except one were in the relapsed or refractory cohort. Two deaths were deemed treatment related (septic shock with 60 mg/m2; pneumonia with 90 mg/m2). INTERPRETATION Guadecitabine was clinically active with acceptable tolerability in patients with intermediate-risk and high-risk myelodysplastic syndromes. Responses and overall survival in the relapsed or refractory cohort offer the potential of a new therapeutic option for patients for whom currently available hypomethylating agents are not successful. We therefore recommend guadecitabine at a dose of 60 mg/m2 on a 5-day schedule for these patients. FUNDING Astex Pharmaceuticals and Stand Up To Cancer.
Collapse
Affiliation(s)
| | - Gail Roboz
- New York-Presbyterian/Weill Cornell Medical Center, New York, NY, USA
| | | | - Hagop Kantarjian
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ellen Ritchie
- New York-Presbyterian/Weill Cornell Medical Center, New York, NY, USA
| | | | - Casey O'Connell
- Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | | | - Scott Lunin
- Fort Myers Cancer Center, Fort Myers, FL, USA
| | - Todd Rosenblat
- Columbia University Irving Medical Center, New York, NY, USA
| | - Karen Yee
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Wendy Stock
- The University of Chicago Medicine Comprehensive Cancer Center, Chicago, IL, USA
| | | | - Joseph Mace
- Florida Cancer Specialists & Research Institute, St Petersburg, FL, USA
| | | | | | - Elias Jabbour
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jean-Pierre J Issa
- Fels Institute for Cancer Research & Molecular Biology, Temple University, Philadelphia, PA, USA
| | - Yong Hao
- Astex Pharmaceuticals, Pleasanton, CA, USA
| | | | | | - Michael R Savona
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
21
|
Lamble AJ, Lind EF. Targeting the Immune Microenvironment in Acute Myeloid Leukemia: A Focus on T Cell Immunity. Front Oncol 2018; 8:213. [PMID: 29951373 PMCID: PMC6008423 DOI: 10.3389/fonc.2018.00213] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 05/24/2018] [Indexed: 12/27/2022] Open
Abstract
Immunotherapies, such as chimeric antigen receptor T cells, bispecific antibodies, and immune checkpoint inhibitors, have emerged as promising modalities in multiple hematologic malignancies. Despite the excitement surrounding immunotherapy, it is currently not possible to predict which patients will respond. Within solid tumors, the status of the immune microenvironment provides valuable insight regarding potential responses to immune therapies. Much less is known about the immune microenvironment within hematologic malignancies but the characteristics of this environment are likely to serve a similar predictive role. Acute myeloid leukemia (AML) is the most common hematologic malignancy in adults, and only 25% of patients are alive 5 years following their diagnosis. There is evidence that manipulation of the immune microenvironment by leukemia cells may play a role in promoting therapy resistance and disease relapse. In addition, it has long been documented that through modulation of the immune system following allogeneic bone marrow transplant, AML can be cured, even in patients with the highest risk disease. These concepts, along with the poor prognosis associated with this disease, have encouraged many groups to start exploring the utility of novel immune therapies in AML. While the implementation of these therapies into clinical trials for AML has been supported by preclinical rationale, many questions still exist surrounding their efficacy, tolerability, and the overall optimal approach. In this review, we discuss what is known about the immune microenvironment within AML with a specific focus on T cells and checkpoints, along with their implications for immune therapies.
Collapse
Affiliation(s)
- Adam J Lamble
- Pediatric Hematology/Oncology, Seattle Children's Hospital, Seattle, WA, United States
| | - Evan F Lind
- Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
22
|
Li CW, Lai TY, Chen BS. Changes of signal transductivity and robustness of gene regulatory network in the carcinogenesis of leukemic subtypes via microarray sample data. Oncotarget 2018; 9:23636-23660. [PMID: 29805763 PMCID: PMC5955113 DOI: 10.18632/oncotarget.25318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 04/11/2018] [Indexed: 11/25/2022] Open
Abstract
Mutation accumulation and epigenetic alterations in genes are important for carcinogenesis. Because leukemogenesis-related signal pathways have been investigated and microarray sample data have been produced in acute myeloid leukemia (AML), myelodysplastic syndromes (MDS) and normal cells, systems analysis in coupling pathways becomes possible. Based on system modeling and identification, we could construct the coupling pathways and their associated gene regulatory networks using microarray sample data. By applying system theory to the estimated system model in coupling pathways, we can then obtain transductivity sensitivity, basal sensitivity and error sensitivity of each protein to identify the potential impact of genetic mutations, epigenetic alterations and the coupling of other pathways from the perspective of energy, respectively. By comparing the results in AML, MDS and normal cells, we investigated the potential critical genetic mutations and epigenetic alterations that activate or repress specific cellular functions to promote MDS or AML leukemogenesis. We suggested that epigenetic modification of β-catenin and signal integration of CSLs, AP-2α, STATs, c-Jun and β-catenin could contribute to cell proliferation at AML and MDS. Epigenetic regulation of ERK and genetic mutation of p53 could lead to the repressed apoptosis, cell cycle arrest and DNA repair in leukemic cells. Genetic mutation of JAK, epigenetic regulation of ERK, and signal integration of C/EBPα could result in the promotion of MDS cell differentiation. According to the results, we proposed three drugs, decitabine, genistein, and monorden for preventing AML leukemogenesis, while three drugs, decitabine, thalidomide, and geldanamycin, for preventing MDS leukemogenesis.
Collapse
Affiliation(s)
- Cheng-Wei Li
- Laboratory of Control and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Tzu-Ying Lai
- Laboratory of Control and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Bor-Sen Chen
- Laboratory of Control and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
23
|
Palomo L, Malinverni R, Cabezón M, Xicoy B, Arnan M, Coll R, Pomares H, García O, Fuster-Tormo F, Grau J, Feliu E, Solé F, Buschbeck M, Zamora L. DNA methylation profile in chronic myelomonocytic leukemia associates with distinct clinical, biological and genetic features. Epigenetics 2018; 13:8-18. [PMID: 29160764 DOI: 10.1080/15592294.2017.1405199] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Chromosomal abnormalities are detected in 20-30% of patients with chronic myelomonocytic leukemia (CMML) and correlate with prognosis. On the mutation level, disruptive alterations are particularly frequent in chromatin regulatory genes. However, little is known about the consequential alterations in the epigenetic marking of the genome. Here, we report the analysis of genomic DNA methylation patterns of 64 CMML patients and 10 healthy controls, using a DNA methylation microarray focused on promoter regions. Differential methylation analysis between patients and controls allowed us to identify abnormalities in DNA methylation, including hypermethylation of specific genes and large genome regions with aberrant DNA methylation. Unsupervised hierarchical cluster analysis identified two main clusters that associated with the clinical, biological, and genetic features of patients. Group 1 was enriched in patients with adverse clinical and biological characteristics and poorer overall and progression-free survival. In addition, significant differences in DNA methylation were observed between patients with low risk and intermediate/high risk karyotypes and between TET2 mutant and wild type patients. Taken together, our results demonstrate that altered DNA methylation patterns reflect the CMML disease state and allow to identify patient groups with distinct clinical features.
Collapse
Affiliation(s)
- Laura Palomo
- a MDS Group. Josep Carreras Leukaemia Research Institute (IJC), ICO-Hospital Germans Trias i Pujol , Universitat Autònoma de Barcelona , Carretera de Can Ruti, Camí de les Escoles, s/n. 08916, Badalona ( Barcelona ), Spain.,b Departament de Bioquímica i Biologia Molecular , Universitat Autònoma de Barcelona , Campus de la UAB, Plaça Cívica, s/n. 08913, Bellaterra ( Barcelona ), Spain
| | - Roberto Malinverni
- c Chromatin, Metabolism and Cell Fate Group. Josep Carreras Leukaemia Research Institute (IJC), Campus ICO-Hospital Germans Trias i Pujol , Program for Predictive and Personalized Medicine of Cancer at the Institute Germans Trias i Pujol (PMPPC-IGTP) , Carretera de Can Ruti, Camí de les Escoles, s/n. 08916, Badalona ( Barcelona ), Spain
| | - Marta Cabezón
- d Hematology Service, ICO-Hospital Germans Trias i Pujol, Josep Carreras Leukaemia Research Institute (IJC) , Universitat Autònoma de Barcelona , Carretera del Canyet, s/n. 08916, Badalona ( Barcelona ), Spain
| | - Blanca Xicoy
- d Hematology Service, ICO-Hospital Germans Trias i Pujol, Josep Carreras Leukaemia Research Institute (IJC) , Universitat Autònoma de Barcelona , Carretera del Canyet, s/n. 08916, Badalona ( Barcelona ), Spain
| | - Montserrat Arnan
- e Hematology Service , ICO-Hospital Duran i Reynals , Avinguda de la Gran Via de l'Hospitalet, 199-203, 08908 Hospitalet de Llobregat ( Barcelona ), Spain
| | - Rosa Coll
- f Hematology Service , ICO-Girona Hospital Josep Trueta, Girona, Spain , Avenida França, s/n. 17007 Girona , Spain
| | - Helena Pomares
- e Hematology Service , ICO-Hospital Duran i Reynals , Avinguda de la Gran Via de l'Hospitalet, 199-203, 08908 Hospitalet de Llobregat ( Barcelona ), Spain
| | - Olga García
- d Hematology Service, ICO-Hospital Germans Trias i Pujol, Josep Carreras Leukaemia Research Institute (IJC) , Universitat Autònoma de Barcelona , Carretera del Canyet, s/n. 08916, Badalona ( Barcelona ), Spain
| | - Francisco Fuster-Tormo
- a MDS Group. Josep Carreras Leukaemia Research Institute (IJC), ICO-Hospital Germans Trias i Pujol , Universitat Autònoma de Barcelona , Carretera de Can Ruti, Camí de les Escoles, s/n. 08916, Badalona ( Barcelona ), Spain
| | - Javier Grau
- d Hematology Service, ICO-Hospital Germans Trias i Pujol, Josep Carreras Leukaemia Research Institute (IJC) , Universitat Autònoma de Barcelona , Carretera del Canyet, s/n. 08916, Badalona ( Barcelona ), Spain
| | - Evarist Feliu
- d Hematology Service, ICO-Hospital Germans Trias i Pujol, Josep Carreras Leukaemia Research Institute (IJC) , Universitat Autònoma de Barcelona , Carretera del Canyet, s/n. 08916, Badalona ( Barcelona ), Spain
| | - Francesc Solé
- a MDS Group. Josep Carreras Leukaemia Research Institute (IJC), ICO-Hospital Germans Trias i Pujol , Universitat Autònoma de Barcelona , Carretera de Can Ruti, Camí de les Escoles, s/n. 08916, Badalona ( Barcelona ), Spain
| | - Marcus Buschbeck
- c Chromatin, Metabolism and Cell Fate Group. Josep Carreras Leukaemia Research Institute (IJC), Campus ICO-Hospital Germans Trias i Pujol , Program for Predictive and Personalized Medicine of Cancer at the Institute Germans Trias i Pujol (PMPPC-IGTP) , Carretera de Can Ruti, Camí de les Escoles, s/n. 08916, Badalona ( Barcelona ), Spain
| | - Lurdes Zamora
- d Hematology Service, ICO-Hospital Germans Trias i Pujol, Josep Carreras Leukaemia Research Institute (IJC) , Universitat Autònoma de Barcelona , Carretera del Canyet, s/n. 08916, Badalona ( Barcelona ), Spain
| |
Collapse
|
24
|
Cull AH, Snetsinger B, Buckstein R, Wells RA, Rauh MJ. Tet2 restrains inflammatory gene expression in macrophages. Exp Hematol 2017; 55:56-70.e13. [PMID: 28826859 DOI: 10.1016/j.exphem.2017.08.001] [Citation(s) in RCA: 214] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 08/07/2017] [Accepted: 08/08/2017] [Indexed: 12/13/2022]
Abstract
Tet methylcytosine dioxygenase 2 (TET2) is one of the earliest and most frequently mutated genes in clonal hematopoiesis of indeterminate potential (CHIP) and myeloid cancers, including myelodysplastic syndromes (MDS) and chronic myelomonocytic leukemia (CMML). TET2 catalyzes the oxidation of 5-methylcytosine to 5-hydroxymethylcytosine, leading to DNA demethylation, and also affects transcription by recruiting histone modifiers. Inactivating TET2 mutations cause epigenetic dysregulation, clonal hematopoietic stem cell (HSC) dominance, and monocytic lineage skewing. Here, we found that Tet2 was the most highly expressed Tet enzyme in murine macrophage (MΦ) differentiation. Tet2 transcription was further induced by lipopolysaccharide (LPS), but not interleukin (IL)-4, stimulation, potentially in a nuclear factor κβ-dependent manner. Tet2 loss did not affect early LPS gene responses in vitro, but increased Il-1b, Il-6, and Arginase 1 (Arg1) mRNA expression at later stages of stimulation in bone-marrow-derived MΦs (BMMΦs). Tet2-deficient peritoneal MΦs, however, demonstrated profound, constitutive expression of LPS-induced genes associated with an inflammatory state in vivo. In contrast, Tet2 deficiency did not affect alternative MΦ gene expression significantly in response to IL-4. These results suggested impaired resolution of inflammation in the absence of Tet2 both in vitro and in vivo. For the first time, we also detected TET2 mutations in BMMΦs from MDS and CMML patients and assayed their effects on LPS responses, including their potential influence on human IL-6 expression. Our results show that Tet2 restrains inflammation in murine MΦs and mice, raising the possibility that loss of TET2 function in MΦs may alter the immune environment in the large elderly population with TET2-mutant CHIP and in TET2-mutant myeloid cancer patients.
Collapse
Affiliation(s)
- Alyssa H Cull
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Brooke Snetsinger
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Rena Buckstein
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Richard A Wells
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Michael J Rauh
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
25
|
Shenoy N, Bhagat T, Nieves E, Stenson M, Lawson J, Choudhary GS, Habermann T, Nowakowski G, Singh R, Wu X, Verma A, Witzig TE. Upregulation of TET activity with ascorbic acid induces epigenetic modulation of lymphoma cells. Blood Cancer J 2017; 7:e587. [PMID: 28731456 PMCID: PMC5549257 DOI: 10.1038/bcj.2017.65] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 06/20/2017] [Indexed: 12/20/2022] Open
Abstract
The Ten Eleven Translocation (TET) enzymes have been found to be mutated in both diffuse large B-cell (DLBCL) and peripheral T-cell (PTCL) lymphomas resulting in DNA hypermethylation. Recent studies in embryonal stem cells showed that ascorbic acid (AA) is a cofactor for TET with a binding site at the catalytic domain, and enhances TET activity. We hypothesized that AA could potentially enhance TET activity in lymphoma cells to cause DNA demethylation, reactivate expression of tumor suppressor genes and enhance chemosensitivity. We demonstrate in vitro that AA treatment of DLBCL and PTCL cells using AA concentrations achievable intravenously increased TET activity leading to DNA demethylation. This epigenetic effect is independent of hydrogen peroxide. AA treatment increased the expression of SMAD1, a tumor suppressor gene known to be suppressed by methylation, and increased chemosensitivity of lymphoma cells. Twenty-nine percent (10/34) of unselected lymphoma patients had plasma AA levels that were deficient suggesting an additional clinical mechanism of TET hypofunction. These data indicate that AA has the potential to modify TET function in lymphoma and enhance chemosensitivity. In addition, the AA deficiency seen in some patients may further impair TET function and contribute to resistance. Clinical trials testing intravenous AA with chemotherapy are warranted.
Collapse
Affiliation(s)
- N Shenoy
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - T Bhagat
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - E Nieves
- Biochemistry and Developmental & Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - M Stenson
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - J Lawson
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - G S Choudhary
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - T Habermann
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - G Nowakowski
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - R Singh
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - X Wu
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - A Verma
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - T E Witzig
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
26
|
Garcia-Manero G, Sekeres MA, Egyed M, Breccia M, Graux C, Cavenagh JD, Salman H, Illes A, Fenaux P, DeAngelo DJ, Stauder R, Yee K, Zhu N, Lee JH, Valcarcel D, MacWhannell A, Borbenyi Z, Gazi L, Acharyya S, Ide S, Marker M, Ottmann OG. A phase 1b/2b multicenter study of oral panobinostat plus azacitidine in adults with MDS, CMML or AML with ⩽30% blasts. Leukemia 2017; 31:2799-2806. [PMID: 28546581 PMCID: PMC5729337 DOI: 10.1038/leu.2017.159] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 02/13/2017] [Accepted: 02/23/2017] [Indexed: 12/15/2022]
Abstract
Treatment with azacitidine (AZA), a demethylating agent, prolonged overall survival (OS) vs conventional care in patients with higher-risk myelodysplastic syndromes (MDS). As median survival with monotherapy is <2 years, novel agents are needed to improve outcomes. This phase 1b/2b trial (n=113) was designed to determine the maximum tolerated dose (MTD) or recommended phase 2 dose (RP2D) of panobinostat (PAN)+AZA (phase 1b) and evaluate the early efficacy and safety of PAN+AZA vs AZA monotherapy (phase 2b) in patients with higher-risk MDS, chronic myelomonocytic leukemia or oligoblastic acute myeloid leukemia with <30% blasts. The MTD was not reached; the RP2D was PAN 30 mg plus AZA 75 mg/m2. More patients receiving PAN+AZA achieved a composite complete response ([CR)+morphologic CR with incomplete blood count+bone marrow CR (27.5% (95% CI, 14.6–43.9%)) vs AZA (14.3% (5.4–28.5%)). However, no significant difference was observed in the 1-year OS rate (PAN+AZA, 60% (50–80%); AZA, 70% (50–80%)) or time to progression (PAN+AZA, 70% (40–90%); AZA, 70% (40–80%)). More grade 3/4 adverse events (97.4 vs 81.0%) and on-treatment deaths (13.2 vs 4.8%) occurred with PAN+AZA. Further dose or schedule optimization may improve the risk/benefit profile of this regimen.
Collapse
Affiliation(s)
- G Garcia-Manero
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M A Sekeres
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, USA
| | - M Egyed
- Kaposi Mor County Teaching Hospital, Kasposvár, Hungary
| | | | - C Graux
- Mont-Godinne University Hospital, Yvoir, Belgium
| | | | - H Salman
- Augusta University, Augusta, GA, USA
| | - A Illes
- University of Debrecen, Debrecen, Hungary
| | - P Fenaux
- Hôpital Saint-Louis, Université Paris Diderot, Paris, France
| | | | - R Stauder
- Innsbruck Medical University, Innsbruck, Austria
| | - K Yee
- Princess Margaret Cancer Centre, Toronto, Canada
| | - N Zhu
- University of Alberta Hospital, Edmonton, Canada
| | - J-H Lee
- Asan Medical Center, University of Ulsan, Seoul, South Korea
| | | | - A MacWhannell
- The Royal Wolverhampton Hospitals NHS Trust, Wolverhampton, UK
| | | | - L Gazi
- Novartis Pharma AG, Basel, Switzerland
| | - S Acharyya
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - S Ide
- Novartis Pharmaceuticals Corporation, Cambridge, MA, USA
| | - M Marker
- Novartis Pharma S.A.S., Rueil-Malmaison, France
| | - O G Ottmann
- Department of Haematology, School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
27
|
Over-expression of miR-196b-5p is significantly associated with the progression of myelodysplastic syndrome. Int J Hematol 2017; 105:777-783. [PMID: 28224273 DOI: 10.1007/s12185-017-2201-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 02/14/2017] [Accepted: 02/14/2017] [Indexed: 12/14/2022]
Abstract
Myelodysplastic syndrome (MDS) is a clonal stem cell disorder characterized by ineffective hematopoiesis with a high risk of transformation to acute myeloid leukemia (AML). miRNAs function as tumor suppressors and oncogenes in various cancers and regulate the differentiation potential of hematopoietic stem and progenitor cells (HSPCs). It has been suggested that miRNAs may play an important role in progression of MDS. We analyzed bone marrow samples collected from MDS patients according to different risk stratification indicated by the International Prognostic Scoring System (IPSS). We demonstrated that miR-196b-5p was up-regulated in intermediate II and higher groups, and in secondary AML (s-AML) patients in particular (P < 0.01) compared with healthy controls, suggesting that the higher expression levels are associated with increased risk of the development of MDS. We observed changes in proliferation and apoptosis in MDS-L cells following transfection with miR-196-5p mimics or inhibitors. After up-regulating the expression of miR-196b-5p, proliferation of MDS-L cells was up-regulated, whereas apoptosis was down-regulated (P < 0.05). In contrast, down-regulation of miR-196b-5p expression decreased cell proliferation and increased apoptosis (P < 0.05). We concluded that over-expression of miR-196b-5p may be closely associated with the risk of transformation to leukemia in MDS patients.
Collapse
|
28
|
Vey N, Prebet T, Thalamas C, Charbonnier A, Rey J, Kloos I, Liu E, Luan Y, Vezan R, Graef T, Recher C. Phase 1 dose-escalation study of oral abexinostat for the treatment of patients with relapsed/refractory higher-risk myelodysplastic syndromes, acute myeloid leukemia, or acute lymphoblastic leukemia. Leuk Lymphoma 2016; 58:1880-1886. [PMID: 27911138 DOI: 10.1080/10428194.2016.1263843] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Histone deacetylase (HDAC) inhibitor abexinostat is under investigation for the treatment of various cancers. Epigenetic changes including aberrant HDAC activity are associated with cancers, including myelodysplastic syndromes (MDS), acute myeloid leukemia (AML), and acute lymphoblastic leukemia (ALL). In this phase 1 dose-escalation study, 17 patients with relapsed/refractory higher-risk MDS, AML, or ALL received oral abexinostat (60, 80 [starting dose], 100, or 120 mg) twice daily (bid) on Days 1-14 of 21-day cycles. The most common treatment-related grade ≥3 adverse events were thrombocytopenia (29%) and neutropenia (24%), none of which led to discontinuation. Maximum-tolerated dose was not reached. Of 12 evaluable patients, best response was stable disease in 1 patient. This study was closed due to limited clinical benefit. Future development of oral abexinostat 100 mg bid in patients with MDS, AML, or ALL should focus on combination regimens. ISRCTN registry: 99680465.
Collapse
Affiliation(s)
- Norbert Vey
- a Department of Hematology , Institut Paoli Calmettes, SIRIC-Marseille , Marseille , France.,b Aix-Marseille University , Marseille , France
| | - Thomas Prebet
- c Internal Medicine Department, Section of Hematology , Yale Cancer Center at Yale University , New Haven , CT , USA
| | - Claire Thalamas
- d Department of Medical Pharmacology , CIC 1436, Université Toulouse Hospital, Inserm , Toulouse , France
| | - Aude Charbonnier
- a Department of Hematology , Institut Paoli Calmettes, SIRIC-Marseille , Marseille , France
| | - Jerome Rey
- a Department of Hematology , Institut Paoli Calmettes, SIRIC-Marseille , Marseille , France
| | - Ioana Kloos
- e Department of Oncology Research and Development , Institut de Recherches Internationales Servier , Suresnes , France
| | - Emily Liu
- f Biometrics, Pharmacyclics , LLC, an AbbVie Company , Sunnyvale , CA , USA
| | - Ying Luan
- f Biometrics, Pharmacyclics , LLC, an AbbVie Company , Sunnyvale , CA , USA
| | - Remus Vezan
- g Department of Clinical Science, Pharmacyclics , LLC, an AbbVie Company , Sunnyvale , CA , USA
| | - Thorsten Graef
- g Department of Clinical Science, Pharmacyclics , LLC, an AbbVie Company , Sunnyvale , CA , USA
| | - Christian Recher
- h Service d'Hématologie , Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole , Toulouse , France.,i Cancer Research Center of Toulouse, UMR1037-INSERM, ERL5294-CNRS , Toulouse , France
| |
Collapse
|
29
|
Jiang X, Wang Z, Ding B, Yin C, Zhong Q, Carter BZ, Yu G, Jiang L, Ye J, Dai M, Zhang Y, Liang S, Zhao Q, Liu Q, Meng F. The hypomethylating agent decitabine prior to chemotherapy improves the therapy efficacy in refractory/relapsed acute myeloid leukemia patients. Oncotarget 2016; 6:33612-22. [PMID: 26384351 PMCID: PMC4741789 DOI: 10.18632/oncotarget.5600] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 08/26/2015] [Indexed: 12/16/2022] Open
Abstract
In this study, we investigated the effect of pre-treatment with demethylating agent decitabine on susceptibility to chemotherapeutic drugs in HL60/ADR, Kasumi-1 and primary AML cells. Cytotoxic effect was increased by decitabine through activation of p53 and inhibition of c-Myc, Survivin and Bcl-2. We demonstrated in clinic that combination of decitabine and HAA consisting of harringtonine, aclarubicin and cytarabine was effective and safe to treat patients with refractory, relapsed or high-risk AML. Decitabine prior to HAA regimen improved the first induction complete response rate, and significantly prolonged overall survival and disease-free survival in these patients compared with HAA alone. These findings support clinic protocols based on decitabine prior to chemotherapy to overcome resistance and improve therapeutic efficacy in AML patients.
Collapse
Affiliation(s)
- Xuejie Jiang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhixiang Wang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bingjie Ding
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Changxin Yin
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qingxiu Zhong
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Hematopathy Diagnosis and Therapy Center, Kanghua Hospital, Dongguan, China
| | - Bing Z Carter
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Guopan Yu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ling Jiang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jieyu Ye
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Min Dai
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yu Zhang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | | | - Qingxia Zhao
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fanyi Meng
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Hematopathy Diagnosis and Therapy Center, Kanghua Hospital, Dongguan, China
| |
Collapse
|
30
|
Iron-induced epigenetic abnormalities of mouse bone marrow through aberrant activation of aconitase and isocitrate dehydrogenase. Int J Hematol 2016; 104:491-501. [DOI: 10.1007/s12185-016-2054-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 06/24/2016] [Accepted: 06/24/2016] [Indexed: 12/19/2022]
|
31
|
Mutations of myelodysplastic syndromes (MDS): An update. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2016; 769:47-62. [DOI: 10.1016/j.mrrev.2016.04.009] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/11/2016] [Indexed: 01/08/2023]
|
32
|
The nucleotidohydrolases DCTPP1 and dUTPase are involved in the cellular response to decitabine. Biochem J 2016; 473:2635-43. [PMID: 27325794 DOI: 10.1042/bcj20160302] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 06/17/2016] [Indexed: 02/03/2023]
Abstract
Decitabine (5-aza-2'-deoxycytidine, aza-dCyd) is an anti-cancer drug used clinically for the treatment of myelodysplastic syndromes and acute myeloid leukaemia that can act as a DNA-demethylating or genotoxic agent in a dose-dependent manner. On the other hand, DCTPP1 (dCTP pyrophosphatase 1) and dUTPase are two 'house-cleaning' nucleotidohydrolases involved in the elimination of non-canonical nucleotides. In the present study, we show that exposure of HeLa cells to decitabine up-regulates the expression of several pyrimidine metabolic enzymes including DCTPP1, dUTPase, dCMP deaminase and thymidylate synthase, thus suggesting their contribution to the cellular response to this anti-cancer nucleoside. We present several lines of evidence supporting that, in addition to the formation of aza-dCTP (5-aza-2'-deoxycytidine-5'-triphosphate), an alternative cytotoxic mechanism for decitabine may involve the formation of aza-dUMP, a potential thymidylate synthase inhibitor. Indeed, dUTPase or DCTPP1 down-regulation enhanced the cytotoxic effect of decitabine producing an accumulation of nucleoside triphosphates containing uracil as well as uracil misincorporation and double-strand breaks in genomic DNA. Moreover, DCTPP1 hydrolyses the triphosphate form of decitabine with similar kinetic efficiency to its natural substrate dCTP and prevents decitabine-induced global DNA demethylation. The data suggest that the nucleotidohydrolases DCTPP1 and dUTPase are factors involved in the mode of action of decitabine with potential value as enzymatic targets to improve decitabine-based chemotherapy.
Collapse
|
33
|
|
34
|
H2AX deficiency is associated with erythroid dysplasia and compromised haematopoietic stem cell function. Sci Rep 2016; 6:19589. [PMID: 26791933 PMCID: PMC4726203 DOI: 10.1038/srep19589] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 11/10/2015] [Indexed: 12/22/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are clonal disorders of haematopoiesis characterised by dysplastic changes of major myeloid cell lines. However, the mechanisms underlying these dysplastic changes are poorly understood. Here, we used a genetically modified mouse model and human patient data to examine the physiological roles of H2AX in haematopoiesis and how the loss of H2AX contributes to dyserythropoiesis in MDS. H2AX knockout mice showed cell-autonomous anaemia and erythroid dysplasia, mimicking dyserythropoiesis in MDS. Also, dyserythropoiesis was increased in MDS patients with the deletion of chromosome 11q23, where H2AX is located. Although loss of H2AX did not affect the early stage of terminal erythropoiesis, enucleation was decreased. H2AX deficiency also led to the loss of quiescence of hematopoietic stem and progenitor cells, which dramatically compromised their bone marrow engraftment. These results reveal important roles of H2AX in late-stage terminal erythropoiesis and hematopoietic stem cell function.
Collapse
|
35
|
Miltiades P, Lamprianidou E, Vassilakopoulos TP, Papageorgiou SG, Galanopoulos AG, Kontos CK, Adamopoulos PG, Nakou E, Vakalopoulou S, Garypidou V, Papaioannou M, Hatjiharissi E, Papadaki HA, Spanoudakis E, Pappa V, Scorilas A, Tsatalas C, Kotsianidis I. The Stat3/5 Signaling Biosignature in Hematopoietic Stem/Progenitor Cells Predicts Response and Outcome in Myelodysplastic Syndrome Patients Treated with Azacitidine. Clin Cancer Res 2015; 22:1958-68. [PMID: 26700206 DOI: 10.1158/1078-0432.ccr-15-1288] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 11/26/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Azacitidine is the mainstay of high-risk myelodysplastic syndromes (MDS) therapy, but molecular predictors of response and the mechanisms of resistance to azacitidine remain largely unidentified. Deregulation of signaling via Stat3 and Stat5 in acute myeloid leukemia (AML) is associated with aggressive disease. Numerous genes involved in cell signaling are aberrantly methylated in MDS, yet the alterations and the effect of azacitidine treatment on Stat3/5 signaling in high-risk MDS have not been explored. EXPERIMENTAL DESIGN We assessed longitudinally constitutive and ligand-induced phospho-Stat3/5 signaling responses by multiparametric flow cytometry in 74 patients with MDS and low blast count AML undergoing azacitidine therapy. Pretreatment Stat3/5 signaling profiles in CD34(+)cells were grouped by unsupervised clustering. The differentiation stage and the molecular properties of the CD34(+)G-CSF-inducible Stat3/5 double-positive subpopulation were performed by flow cytometry and quantitative real-time PCR in isolated MDS progenitors. RESULTS The pretreatment Stat3/5 signaling profiles in CD34(+)cells correlated strongly with response and cytogenetics and independently predicted event-free survival. We further identified a CD34(+)G-CSF-inducible Stat3/5 double-positive subpopulation (DP subset) whose pretreatment levels were inversely associated with treatment response and cytogenetics. The kinetics of the DP subset followed the response to azacitidine and the disease course, whereas its molecular characteristics and cellular hierarchy were consistent with a leukemia propagating cell phenotype. CONCLUSIONS Our findings provide a novel link among Stat3/5 signaling and MDS pathobiology and suggest that the Stat3/5 signaling biosignature may serve as both a response biomarker and treatment target.
Collapse
Affiliation(s)
- Paraskevi Miltiades
- Department of Hematology, Democritus University of Thrace, Alexandroupolis, Greece
| | | | - Theodoros P Vassilakopoulos
- Department of Hematology, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Sotirios G Papageorgiou
- Second Department of Internal Medicine, Hematology Unit, Attikon University General Hospital, Athens, Greece
| | | | - Christos K Kontos
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis G Adamopoulos
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelia Nakou
- Department of Hematology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Sofia Vakalopoulou
- Second Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration Hospital, Thessaloniki, Greece
| | - Vassilia Garypidou
- Second Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration Hospital, Thessaloniki, Greece
| | - Maria Papaioannou
- Department of Haematology, Aristotle University of Thessaloniki, AHEPA Hospital, Thessaloniki, Greece
| | - Evdoxia Hatjiharissi
- Department of Hematology, Theageneion Hospital of Thessaloniki, Thessaloniki, Greece
| | - Helen A Papadaki
- Department of Hematology, University Hospital of Heraklion, Heraklion, Greece
| | - Emmanuil Spanoudakis
- Department of Hematology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Vassiliki Pappa
- Second Department of Internal Medicine, Hematology Unit, Attikon University General Hospital, Athens, Greece
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Ioannis Kotsianidis
- Department of Hematology, Democritus University of Thrace, Alexandroupolis, Greece.
| | | |
Collapse
|
36
|
Belo H, Silva G, Cardoso BA, Porto B, Minguillon J, Barbot J, Coutinho J, Casado JA, Benedito M, Saturnino H, Costa E, Bueren JA, Surralles J, Almeida A. Epigenetic Alterations in Fanconi Anaemia: Role in Pathophysiology and Therapeutic Potential. PLoS One 2015; 10:e0139740. [PMID: 26466379 PMCID: PMC4605638 DOI: 10.1371/journal.pone.0139740] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 09/15/2015] [Indexed: 01/23/2023] Open
Abstract
Fanconi anaemia (FA) is an inherited disorder characterized by chromosomal instability. The phenotype is variable, which raises the possibility that it may be affected by other factors, such as epigenetic modifications. These play an important role in oncogenesis and may be pharmacologically manipulated. Our aim was to explore whether the epigenetic profiles in FA differ from non-FA individuals and whether these could be manipulated to alter the disease phenotype. We compared expression of epigenetic genes and DNA methylation profile of tumour suppressor genes between FA and normal samples. FA samples exhibited decreased expression levels of genes involved in epigenetic regulation and hypomethylation in the promoter regions of tumour suppressor genes. Treatment of FA cells with histone deacetylase inhibitor Vorinostat increased the expression of DNM3Tβ and reduced the levels of CIITA and HDAC9, PAK1, USP16, all involved in different aspects of epigenetic and immune regulation. Given the ability of Vorinostat to modulate epigenetic genes in FA patients, we investigated its functional effects on the FA phenotype. This was assessed by incubating FA cells with Vorinostat and quantifying chromosomal breaks induced by DNA cross-linking agents. Treatment of FA cells with Vorinostat resulted in a significant reduction of aberrant cells (81% on average). Our results suggest that epigenetic mechanisms may play a role in oncogenesis in FA. Epigenetic agents may be helpful in improving the phenotype of FA patients, potentially reducing tumour incidence in this population.
Collapse
Affiliation(s)
- Hélio Belo
- Unidade de Investigação em Patobiologia Molecular, Instituto Português de Oncologia de Lisboa Francisco Gentil, E.P.E., Lisboa, Portugal
- CEDOC, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Gabriela Silva
- Unidade de Investigação em Patobiologia Molecular, Instituto Português de Oncologia de Lisboa Francisco Gentil, E.P.E., Lisboa, Portugal
- CEDOC, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Bruno A. Cardoso
- Unidade de Investigação em Patobiologia Molecular, Instituto Português de Oncologia de Lisboa Francisco Gentil, E.P.E., Lisboa, Portugal
- CEDOC, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Beatriz Porto
- Laboratório de Citogenética do Instituto de Ciências Biomédicas de Abel Salazar, Porto, Portugal
| | - Jordi Minguillon
- Center for Biomedical Network Research on Rare Diseases (CIBERER) and Department of Genetics and Microbiology, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - José Barbot
- Unidade de Hematologia Pediátrica do Centro Hospitalar do Porto, Porto, Portugal
| | - Jorge Coutinho
- Unidade de Hematologia Pediátrica do Centro Hospitalar do Porto, Porto, Portugal
| | - Jose A. Casado
- Hematopoiesis and Gene Therapy Division, CIEMAT, Madrid, Spain
| | - Manuela Benedito
- Serviço de hematologia do Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Hema Saturnino
- Serviço de hematologia do Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Emília Costa
- Unidade de Hematologia Pediátrica do Centro Hospitalar do Porto, Porto, Portugal
| | - Juan A. Bueren
- Unidade de Hematologia Pediátrica do Centro Hospitalar do Porto, Porto, Portugal
| | - Jordi Surralles
- Center for Biomedical Network Research on Rare Diseases (CIBERER) and Department of Genetics and Microbiology, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Antonio Almeida
- Unidade de Investigação em Patobiologia Molecular, Instituto Português de Oncologia de Lisboa Francisco Gentil, E.P.E., Lisboa, Portugal
- CEDOC, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
- * E-mail:
| |
Collapse
|
37
|
Helbo AS, Treppendahl M, Aslan D, Dimopoulos K, Nandrup-Bus C, Holm MS, Andersen MK, Liang G, Kristensen LS, Grønbæk K. Hypermethylation of the VTRNA1-3 Promoter is Associated with Poor Outcome in Lower Risk Myelodysplastic Syndrome Patients. Genes (Basel) 2015; 6:977-90. [PMID: 26473932 PMCID: PMC4690025 DOI: 10.3390/genes6040977] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 09/09/2015] [Accepted: 09/22/2015] [Indexed: 12/13/2022] Open
Abstract
Myelodysplastic syndrome (MDS) is a heterogeneous group of clonal hematopoietic disorders. MDS is frequently associated with deletions on chromosome 5q as well as aberrant DNA methylation patterns including hypermethylation of key tumor suppressors. We have previously shown that hypermethylation and silencing of the non-coding RNA VTRNA2-1 are correlated with poor outcomes in acute myeloid leukemia patients. In this study, we find that VTRNA1-2 and VTRNA1-3, both located on chromosome 5q, can be regulated and silenced by promoter DNA methylation, and that the hypomethylating agent 5-aza-2-deoxycytidine causes reactivation these genes. In normal hematopoiesis, we find that vault RNAs (vtRNAs) show differential methylation between various hematopoietic cell populations, indicating that allele-specific methylation events may occur during hematopoiesis. In addition, we show that VTRNA1-3 promoter hypermethylation is frequent in lower risk MDS patients and is associated with a decreased overall survival.
Collapse
Affiliation(s)
- Alexandra Søgaard Helbo
- Department of Hematology, Rigshospitalet, University Hospital Copenhagen, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark.
| | - Marianne Treppendahl
- Department of Hematology, Rigshospitalet, University Hospital Copenhagen, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark.
| | - Derya Aslan
- Department of Hematology, Rigshospitalet, University Hospital Copenhagen, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark.
| | - Konstantinos Dimopoulos
- Department of Hematology, Rigshospitalet, University Hospital Copenhagen, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark.
| | - Cecilie Nandrup-Bus
- Department of Hematology, Rigshospitalet, University Hospital Copenhagen, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark.
| | - Mette Skov Holm
- Department of Hematology, Aarhus University Hospital, Tage Hansens Gade 2, 8000 Aarhus C, Denmark.
| | - Mette Klarskov Andersen
- Department of Clinical Genetics, Rigshospitalet, University Hospital Copenhagen, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark.
| | - Gangning Liang
- Department of Biochemistry and Molecular Biology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Los Angeles, CA 90089, USA.
| | - Lasse Sommer Kristensen
- Department of Hematology, Rigshospitalet, University Hospital Copenhagen, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark.
| | - Kirsten Grønbæk
- Department of Hematology, Rigshospitalet, University Hospital Copenhagen, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark.
- Danstem, University of Copenhagen, Blegdamsvej, 2200 Copenhagen N, Denmark.
| |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW This review highlights recent insights into the roles of microRNAs (miRNAs) in pathogenesis of myeloid malignancies and tantalising prospects of miRNA therapy. RECENT FINDINGS New roles for miRNAs in biological and disease processes are constantly being discovered. Although great effort has been put into identifying and cataloguing aberrantly expressed miRNAs in leukaemia, very little is known about the functional consequences of their deregulation in myeloid malignancies. This review will discuss the significance of powerful oncogenic miRNAs such as miR-22 in self-renewal and transformation of haematopoietic stem cells, as well as their ability to induce epigenetic alterations in the pathogenesis of the stem cell disorder myelodysplastic syndromes and myeloid leukaemia. SUMMARY Improved understanding of biological roles of miRNAs in the pathogenesis of haematological malignancies will allow rational stratification of patients and provide new therapeutic entries for the treatment of myelodysplastic syndromes and leukaemia.
Collapse
|
39
|
Byrne M, Bennett RL, Cheng X, May WS. Progressive genomic instability in the Nup98-HoxD13 model of MDS correlates with loss of the PIG-A gene product. Neoplasia 2015; 16:627-33. [PMID: 25220590 PMCID: PMC4234872 DOI: 10.1016/j.neo.2014.07.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 07/09/2014] [Accepted: 07/16/2014] [Indexed: 12/17/2022] Open
Abstract
The Nup98-HoxD13 (NHD13) fusion gene was identified in a patient with therapy-related myelodysplastic syndrome (MDS). When transgenically expressed in hematopoietic cells, mice faithfully recapitulate human disease with serial progression from peripheral blood (PB) cytopenias and increased bone marrow (BM) blasts to acute leukemia. It is well accepted that genomic instability in dysplastic hematopoietic stem/progenitor cells (HSPC) drives the evolution of MDS to acute leukemia. Findings here demonstrate that reticulocytes, myeloid and lymphoid PB cells of NHD13 mice, display an increase in the age-associated loss of glycosylphosphatidylinositol-linked surface proteins versus wild type controls. These data correlate with a progressive increase in the DNA damage response as measured by γ-H2AX activity, accumulating BM blasts as the disease progresses and finally development of acute leukemia. These findings clearly demonstrate a state of progressive genomic instability that increases the likelihood of a “second hit” or complimentary mutation later in the disease to trigger development of acute leukemia and underscores the mechanistic nature of how the NUP98-HoxD13 transgene induces progression of MDS to acute leukemia. Additionally, these data support the use of the PIG-A assay as an efficient, real-time surrogate marker of the genomic instability that occurs in the MDS HSPCs. Key Point The PIG-A assay is a sensitive, nonlethal method for the serial assessment of genomic instability in mouse models of MDS.
Collapse
Affiliation(s)
- Michael Byrne
- Department of Medicine, Division of Hematology and Oncology and the University of Florida Health Cancer Center, Gainesville, FL 32610
| | - Richard L Bennett
- Department of Medicine, Division of Hematology and Oncology and the University of Florida Health Cancer Center, Gainesville, FL 32610
| | - Xiaodong Cheng
- Department of Medicine, Division of Hematology and Oncology and the University of Florida Health Cancer Center, Gainesville, FL 32610
| | - W Stratford May
- Department of Medicine, Division of Hematology and Oncology and the University of Florida Health Cancer Center, Gainesville, FL 32610.
| |
Collapse
|
40
|
Amirizadeh N, Oodi A, Mehrasa R, Nikougoftar M. Apoptosis, DAP-Kinase1 Expression and the Influences of Cytokine Milieu and Mesenchymal Stromal Cells on Ex Vivo Expansion of Umbilical Cord Blood-Derived Hematopoietic Stem Cells. Indian J Hematol Blood Transfus 2015; 32:67-77. [PMID: 26855509 DOI: 10.1007/s12288-015-0545-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 02/05/2014] [Indexed: 10/23/2022] Open
Abstract
Expansion of umbilical cord blood-derived CD34(+) cells can potentially provide them in numbers sufficient for clinical applications in adult humans. In this study apoptosis rate of expanded cells, mRNA expression and promoter methylation status of DAPK1 were evaluated during cord blood hematopoietic stem cell (CB-HSC) ex vivo expansion using cytokines and a co-culture system with mesenchymal stromal cells (MSCs). Ex vivo cultures of CB-HSCs were performed in three culture conditions for 14 days: cytokines with MSCs feeder layer, cytokines without MSCs feeder layer and co-culture with MSCs feeder layer without cytokine. Total number of cells, CD34(+) cells and colony forming unit assay were performed during expansion. Flow cytometric analysis by propidium iodide was performed to detection of apoptosis rate in expanded cells. Methylation status of the DAPK1 gene promoter was analyzed using methylation specific PCR, and DAPK1 mRNA expression was evaluated by real time-PCR. Maximum CB-CD34(+) cells expansion was observed in day 10 of expansion. The highest apoptosis rate was observed in cytokine culture without feeder layer that showed significant difference with co-culture condition. The data showed that ex vivo expansion of CD34(+) cells in all three culture conditions after 10 days resulted in, significant increased expression of DAPK1, also a significant difference between co-culture without cytokine and two other cytokine culture was observed (p < 0.01). DAPK1gene promoter of expanded CD34(+) cells at days 5, 10 and 14 of culture remained in unmethylated form similar to fresh CD34(+) cells. Expression of DAPK1 in hematopoietic cells was increased during 10 days expansion of CD34(+) cells. Also no methylation of DAPK1 promoter was observed; otherwise it would be capable of initiating some leukemic cell progression or disruption in hematopoietic regeneration.
Collapse
Affiliation(s)
- Naser Amirizadeh
- Blood Transfusion Research Center, High Institute for Education and Research in Transfusion Medicine, IBTO Bldg. Hemmat Exp Way, Next to the Milad Tower, P.O. Box 14665-1157, Tehran, Iran
| | - Arezoo Oodi
- Blood Transfusion Research Center, High Institute for Education and Research in Transfusion Medicine, IBTO Bldg. Hemmat Exp Way, Next to the Milad Tower, P.O. Box 14665-1157, Tehran, Iran
| | - Roya Mehrasa
- Blood Transfusion Research Center, High Institute for Education and Research in Transfusion Medicine, IBTO Bldg. Hemmat Exp Way, Next to the Milad Tower, P.O. Box 14665-1157, Tehran, Iran
| | - Mahin Nikougoftar
- Blood Transfusion Research Center, High Institute for Education and Research in Transfusion Medicine, IBTO Bldg. Hemmat Exp Way, Next to the Milad Tower, P.O. Box 14665-1157, Tehran, Iran
| |
Collapse
|
41
|
Peripheral blood mononuclear cell proteome changes in patients with myelodysplastic syndrome. BIOMED RESEARCH INTERNATIONAL 2015; 2015:872983. [PMID: 25969835 PMCID: PMC4415457 DOI: 10.1155/2015/872983] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 03/31/2015] [Indexed: 12/13/2022]
Abstract
Our aim was to search for proteome changes in peripheral blood mononuclear cells (PBMCs) of MDS patients with refractory cytopenia with multilineage dysplasia. PBMCs were isolated from a total of 12 blood samples using a Histopaque-1077 solution. The proteins were fractioned, separated by 2D SDS-PAGE (pI 4–7), and double-stained. The proteomes were compared and statistically processed with Progenesis SameSpots; then proteins were identified by nano-LC-MS/MS. Protein functional association and expression profiles were analyzed using the EnrichNet application and Progenesis SameSpots hierarchical clustering software, respectively. By comparing the cytosolic, membrane, and nuclear fractions of the two groups, 178 significantly (P < 0.05, ANOVA) differing spots were found, corresponding to 139 unique proteins. Data mining of the Reactome and KEGG databases using EnrichNet highlighted the possible involvement of the identified protein alterations in apoptosis, proteasome protein degradation, heat shock protein action, and signal transduction. Western blot analysis revealed underexpression of vinculin and advanced fragmentation of fermitin-3 in MDS patients. To the best of our knowledge, this is the first time that proteome changes have been identified in the mononuclear cells of MDS patients. Vinculin and fermitin-3, the proteins involved in cell adhesion and integrin signaling, have been shown to be dysregulated in MDS.
Collapse
|
42
|
Jabbour E, Garcia-Manero G. Deacetylase inhibitors for the treatment of myelodysplastic syndromes. Leuk Lymphoma 2015; 56:1205-12. [PMID: 25058371 DOI: 10.3109/10428194.2014.946025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Myelodysplastic syndromes (MDS) are a diverse group of myeloid disorders, with patients being at risk for cytopenias or progression to acute myeloid leukemia. Several classification and prognostic scoring systems have been developed. High-intensity treatments are not appropriate for all patients. Two demethylating agents, azacitidine and decitabine, are approved for the treatment of MDS, although many patients do not derive long-term benefit and eventually progress. Deacetylase inhibitors have emerged as novel treatment candidates based on mechanistic rationale and preliminary data. This article reviews existing data on MDS treatment and discusses the rationale and potential for combination with deacetylase inhibitors.
Collapse
Affiliation(s)
- Elias Jabbour
- Leukemia Department, M. D. Anderson Cancer Center , Houston, TX , USA
| | | |
Collapse
|
43
|
Kang H, Wang X, Gao L, Cen J, Li M, Wang W, Wang N, Li Y, Wang L, Yu L. Clinical implications of the quantitative detection of ID4 gene methylation in myelodysplastic syndrome. Eur J Med Res 2015; 20:16. [PMID: 25889027 PMCID: PMC4336702 DOI: 10.1186/s40001-015-0092-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 01/22/2015] [Indexed: 11/29/2022] Open
Abstract
Background Myelodysplastic syndrome (MDS) eventually transforms into acute leukemia (AL) in about 30% of patients. Hypermethylation of the inhibitor of DNA binding 4 (ID4) gene may play an important role in the initiation and development of MDS and AL. The aim of this study was to quantitatively assess ID4 gene methylation in MDS and to establish if it could be an effective method of evaluating MDS disease progression. Methods We examined 142 bone marrow samples from MDS patients, healthy donors and MDS-AL patients using bisulfite sequencing PCR and quantitative real-time methylation-specific PCR. The ID4 methylation rates and levels were assessed. Results ID4 methylation occurred in 27 patients (27/100). ID4 gene methylation was more frequent and at higher levels in patients with advanced disease stages and in high-risk subgroups according to WHO (P < 0.001, P < 0.001, respectively) and International Prognostic Scoring System (IPSS) (P = 0.002, P = 0.007, respectively) classifications. ID4 methylation levels changed during disease progression. Both methylation rates and methylation levels were significantly different between healthy donor, MDS patients and patients with MDS-AL (P < 0.001, P < 0.001, respectively). Multivariate analysis indicated that the level of ID4 methylation was an independent factor influencing overall survival. Patients with MDS showed decreased survival time with increased ID4 methylation levels (P = 0.011, hazard ratio (HR) = 2.371). Patients with ID4 methylation had shorter survival time than those without ID4 methylation (P = 0.008). Conclusions Our findings suggest that ID4 gene methylation might be a new biomarker for MDS monitoring and the detection of minimal residual disease.
Collapse
Affiliation(s)
- Huiyuan Kang
- Department of Hematology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China. .,Department of Clinical Tests, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| | - Xinrong Wang
- Department of Hematology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| | - Li Gao
- Department of Hematology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| | - Jian Cen
- Department of Hematology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| | - Mianyang Li
- Department of Clinical Tests, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| | - Wei Wang
- Department of Hematology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| | - Nan Wang
- Department of Hematology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| | - Yonghui Li
- Department of Hematology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| | - Lili Wang
- Department of Hematology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| | - Li Yu
- Department of Hematology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| |
Collapse
|
44
|
Pellagatti A, Boultwood J. The molecular pathogenesis of the myelodysplastic syndromes. Eur J Haematol 2015; 95:3-15. [DOI: 10.1111/ejh.12515] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2014] [Indexed: 02/07/2023]
Affiliation(s)
- Andrea Pellagatti
- Leukaemia & Lymphoma Research Molecular Haematology Unit; Nuffield Division of Clinical Laboratory Sciences; Radcliffe Department of Medicine; University of Oxford; Oxford UK
| | - Jacqueline Boultwood
- Leukaemia & Lymphoma Research Molecular Haematology Unit; Nuffield Division of Clinical Laboratory Sciences; Radcliffe Department of Medicine; University of Oxford; Oxford UK
| |
Collapse
|
45
|
Abstract
DNA methylation and histone modification are epigenetic mechanisms that result in altered gene expression and cellular phenotype. The exact role of methylation in myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) remains unclear. However, aberrations (e.g. loss-/gain-of-function or up-/down-regulation) in components of epigenetic transcriptional regulation in general, and of the methylation machinery in particular, have been implicated in the pathogenesis of these diseases. In addition, many of these components have been identified as therapeutic targets for patients with MDS/AML, and are also being assessed as potential biomarkers of response or resistance to hypomethylating agents (HMAs). The HMAs 5-azacitidine (AZA) and 2'-deoxy-5-azacitidine (decitabine, DAC) inhibit DNA methylation and have shown significant clinical benefits in patients with myeloid malignancies. Despite being viewed as mechanistically similar drugs, AZA and DAC have differing mechanisms of action. DAC is incorporated 100% into DNA, whereas AZA is incorporated into RNA (80-90%) as well as DNA (10-20%). As such, both drugs inhibit DNA methyltransferases (DNMTs; dependently or independently of DNA replication) resulting in the re-expression of tumor-suppressor genes; however, AZA also has an impact on mRNA and protein metabolism via its inhibition of ribonucleotide reductase, resulting in apoptosis. Herein, we first give an overview of transcriptional regulation, including DNA methylation, post-translational histone-tail modifications, the role of micro-RNA and long-range epigenetic gene silencing. We place special emphasis on epigenetic transcriptional regulation and discuss the implication of various components in the pathogenesis of MDS/AML, their potential as therapeutic targets, and their therapeutic modulation by HMAs and other substances (if known). The main focus of this review is laid on dissecting the rapidly evolving knowledge of AZA and DAC with a special focus on their differing mechanisms of action, and the effect of HMAs on transcriptional regulation.
Collapse
Affiliation(s)
- Lisa Pleyer
- 3rd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory for Immunological and Molecular Cancer Research, Oncologic Center, Paracelsus Medical University Hospital Salzburg, Center for Clinical Cancer and Immunology Trials at Salzburg Cancer Research Institute , Salzburg , Austria
| | | |
Collapse
|
46
|
Abstract
CONTEXT Hematopathology is a dynamic field that has always been on the frontier of clinical research within the scope of pathology. Several recent developments in hematopathology will likely affect its practice clinically. OBJECTIVE To review 5 important recent advances in hematopathology: (1) detection and prognostic implication of MYC in diffuse large B-cell lymphomas, (2) determining origin and prognosis through immunoglobulin gene usage in mature B-cell neoplasms, (3)detecting minimal residual disease in multiple myeloma, (4) using genome-wide analysis in myelodysplastic syndromes, and (5) employing whole-genome sequencing in acute myeloid leukemias. DATA SOURCES Literature review and the authors' experiences in an academic center. CONCLUSIONS These advances will bring hematopathology into a new molecular era and help us to better understand the molecular, pathologic mechanisms of lymphomas, leukemias, myelomas, and myelodysplastic syndromes. They will help us to identify diagnostic and prognostic markers and eventually provide new therapeutic targets and treatments for these diseases.
Collapse
Affiliation(s)
- Min Shi
- From the Department of Pathology, UMass Memorial Medical Center, Worcester, Massachusetts
| | | | | | | |
Collapse
|
47
|
Karlic H, Herrmann H, Varga F, Thaler R, Reitermaier R, Spitzer S, Ghanim V, Blatt K, Sperr WR, Valent P, Pfeilstöcker M. The role of epigenetics in the regulation of apoptosis in myelodysplastic syndromes and acute myeloid leukemia. Crit Rev Oncol Hematol 2014; 90:1-16. [DOI: 10.1016/j.critrevonc.2013.10.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 09/03/2013] [Accepted: 10/02/2013] [Indexed: 01/17/2023] Open
|
48
|
Abstract
The myelodysplastic syndrome (MDS) is a clonal hematologic disorder that frequently evolves to acute myeloid leukemia (AML). Its pathogenesis remains unclear, but mutations in epigenetic modifiers are common and the disease often responds to DNA methylation inhibitors. We analyzed DNA methylation in the bone marrow and spleen in two mouse models of MDS/AML, the NUP98-HOXD13 (NHD13) mouse and the RUNX1 mutant mouse model. Methylation array analysis showed an average of 512/3445 (14.9%) genes hypermethylated in NHD13 MDS, and 331 (9.6%) genes hypermethylated in RUNX1 MDS. Thirty-two percent of genes in common between the two models (2/3 NHD13 mice and 2/3 RUNX1 mice) were also hypermethylated in at least two of 19 human MDS samples. Detailed analysis of 41 genes in mice showed progressive drift in DNA methylation from young to old normal bone marrow and spleen; to MDS, where we detected accelerated age-related methylation; and finally to AML, which markedly extends DNA methylation abnormalities. Most of these genes showed similar patterns in human MDS and AML. Repeat element hypomethylation was rare in MDS but marked the transition to AML in some cases. Our data show consistency in patterns of aberrant DNA methylation in human and mouse MDS and suggest that epigenetically, MDS displays an accelerated aging phenotype.
Collapse
|
49
|
Hascher A, Haase AK, Hebestreit K, Rohde C, Klein HU, Rius M, Jungen D, Witten A, Stoll M, Schulze I, Ogawa S, Wiewrodt R, Tickenbrock L, Berdel WE, Dugas M, Thoennissen NH, Müller-Tidow C. DNA Methyltransferase Inhibition Reverses Epigenetically Embedded Phenotypes in Lung Cancer Preferentially Affecting Polycomb Target Genes. Clin Cancer Res 2013; 20:814-26. [DOI: 10.1158/1078-0432.ccr-13-1483] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
50
|
Lukackova R, Gerykova Bujalkova M, Majerova L, Mladosievicova B. Molecular genetic methods in the diagnosis of myelodysplastic syndromes. A review. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2013; 158:339-45. [PMID: 24263214 DOI: 10.5507/bp.2013.084] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 11/06/2013] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Myelodysplastic syndromes (MDS) represent a heterogeneous group of premalignant hematologic disorders characterized by ineffective hematopoiesis, peripheral blood cytopenias and increased risk of progression to acute leukemia. Cytogenetic analysis still plays a central role in the diagnosis of MDS, as clonal chromosomal abnormalities are observed in 30-50% of MDS patients. Despite their technical limitations, standard karyotyping and fluorescence in situ hybridization (FISH) are routinely used for identifying recurrent chromosomal rearrangements. However, using this approach means that submicroscopic and not targeted chromosomal aberrations, as well as somatic mutations and epigenetic changes remain largely undetected. METHODS AND RESULTS Introduction of methods for the analysis of copy-number variations (CNV), including array-based technologies and Multiplex ligation-dependent probe amplification (MLPA) has provided novel insights into the molecular pathogenesis of MDS and considerably extended possibilities for genetic laboratory testing. Several novel molecular markers have been discovered and used for diagnosis and prognostic evaluation of patients with MDS. At present, mutational analysis is not routinely performed, as the clinical significance of somatic mutations in MDS has only begun to emerge. However, recently introduced Next-generation sequencing (NGS) technologies could help to elucidate the relationship between chromosomal and molecular aberrations in MDS and lead to further improvement in its diagnosis. CONCLUSION This review focuses on the advantages, limitations, clinical applications and future perspectives of three molecular methods (array-based analysis, MLPA and NGS) currently used in genetic testing and/ or translational research of MDS. In conclusion, a brief summary for clinicians from the routine diagnostic point of view is given.
Collapse
Affiliation(s)
- Renata Lukackova
- Department of Clinical Genetics, Medirex a.s., Bratislava, Slovak Republic
| | | | | | | |
Collapse
|