1
|
Zhang N, Li H, Wang D, Wang Z, Zhu JS, Chen K, Jiang H, Shao JB, Cai C. Decitabine as epigenetic priming with CLAG induce improved outcome of relapsed or refractory acute myeloid leukemia in children. Clin Epigenetics 2024; 16:63. [PMID: 38725010 PMCID: PMC11080195 DOI: 10.1186/s13148-024-01677-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Decitabine (DAC), a DNA methyltransferase inhibitor, has shown efficacy combined with chemotherapy for relapsed or refractory (R/R) acute myeloid leukemia (AML) in adults, but less is known about its efficacy in children. Accordingly, we conducted a study which involved a priming regimen consisting of DAC with cladribine, cytarabine, and granulocyte-stimulating factor (DAC-CLAG) and compared the efficacy and safety of this regimen with CLAG alone. METHODS A total of 39 R/R AML children who received the CLAG or DAC-CLAG regimen in Shanghai Children's Hospital were retrospectively enrolled in this non-randomized study. These regimens were studied sequentially over time. Twenty-two patients received CLAG from 2015, while 17 patients were administered epigenetic priming with DAC before CLAG from 2020. Patients were subsequently bridged to stem cell transplantation (SCT) or consolidation chemotherapy. Complete remission (CR) and adverse effects were analyzed by Fisher's exact test, and survival was analyzed by the Kaplan-Meier method. RESULTS DAC-CLAG conferred a numerically higher CR compared to CLAG (70.59% vs 63.64%; P = 0.740). High CR rates occurred in patients with good cytogenetics (P = 0.029) and prior induction without cladribine (P = 0.099). The 1-year event-free survival (EFS) was 64.71% ± 11.59% and 63.31% ± 10.35% in the DAC-CLAG and CLAG group (P = 0.595), and 1-year overall survival (OS) was 81.45% ± 9.72% and 77.01% ± 9.04%, respectively (P = 0.265). The 1-year OS and EFS after SCT were higher in the DAC-CLAG than in the CLAG cohort (100% vs 92.31% ± 7.39%, P = 0.072; 92.31% ± 7.39% vs 85.71% ± 9.35%, P = 0.158). Univariate analysis revealed that a good prognosis included good cytogenetics (P = 0.002), non-complex karyotype (P = 0.056), CR on reinduction (P < 0.0001), and bridging to SCT (P = 0.0007). Use of a hypomethylating agent (P = 0.049) and bridging to SCT (P = 0.011) were independent prognostic factors. Grade 3/4 hematologic toxicity and infection were the main adverse events. CONCLUSIONS DAC prior to the CLAG regimen improved remission in pediatric R/R AML, and was feasible and well tolerated. CLAG ± DAC as a salvage therapy prior to SCT induced improved survival.
Collapse
Affiliation(s)
- Na Zhang
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 1400, West Beijing Road, Shanghai, 200040, China
| | - Hong Li
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 1400, West Beijing Road, Shanghai, 200040, China
| | - Dan Wang
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 1400, West Beijing Road, Shanghai, 200040, China
| | - Zhen Wang
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 1400, West Beijing Road, Shanghai, 200040, China
| | - Jia-Shi Zhu
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 1400, West Beijing Road, Shanghai, 200040, China
| | - Kai Chen
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 1400, West Beijing Road, Shanghai, 200040, China
| | - Hui Jiang
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 1400, West Beijing Road, Shanghai, 200040, China.
| | - Jing-Bo Shao
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 1400, West Beijing Road, Shanghai, 200040, China.
| | - Cheng Cai
- Department of Neonatology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 355, Luding Road, Shanghai, 200062, China.
| |
Collapse
|
2
|
Hui Y, Li Y, Tong X, Huang L, Mao X, Huang L, Zhang D. Reinduction chemotherapy regimen involved decitabine and cladribine improves the prognosis of patients with relapsed or refractory acute myeloid leukemia: A preliminary study. Int J Cancer 2021; 149:901-908. [PMID: 33837553 DOI: 10.1002/ijc.33595] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 11/10/2022]
Abstract
Relapsed/refractory acute myeloid leukemia (R/R-AML) is characterized by a high incidence, short survival and poor prognosis. Presently, no unified effective reinduction chemotherapy regimen has been developed. Therefore, the use of reinduction chemotherapy regimens before allogeneic hematopoietic stem cell transplantation (allo-HSCT) is controversial. Our study aims to analyze the prognostic factors of R/R-AML and to evaluate the efficacy of the regimen involved decitabine, cladribine, idarubicin or homoharringtonine, and cytarabine (DCIA/DCHA). Clinical and survival data of 112 R/R-AML patients were obtained. Among the 102 R/R-AML patients that were treated with conventional regimens, we found that poor prognosis was related to a greater proportion of bone marrow blasts (>70%) and not achieving complete remission (non-CR) after the first reinduction chemotherapy. Hematopoietic stem cell transplantation (of which 89.47% was allo-HSCT) following CR after the first reinduction chemotherapy often improves the prognosis. Of the 10 R/R-AML patients that were treated with the DCIA/DCHA regimen, nine patients achieved CR or complete response with incomplete hematopoietic recovery (CRi) after one course of chemotherapy. The median overall survival of the 10 patients was 10.14 (1.23-29.13) months. In conclusion, non-CR was associated with poor prognosis in R/R-AML. Therefore, intensive reinduction chemotherapy should be selected to achieve CR. This creates conditions for allo-HSCT and improves prognosis of R/R-AML patients. The DCIA/DCHA regimen showed good efficacy and tolerable adverse reactions in R/R-AML treatment. This combination may be used as a bridging regimen for allo-HSCT in R/R-AML.
Collapse
Affiliation(s)
- Yan Hui
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiwen Tong
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lifang Huang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xia Mao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liang Huang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Donghua Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Tobiasson M, Kittang AO. Treatment of myelodysplastic syndrome in the era of next-generation sequencing. J Intern Med 2019; 286:41-62. [PMID: 30869816 DOI: 10.1111/joim.12893] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Next-generation sequencing (NGS) is rapidly changing the clinical care of patients with myelodysplastic syndrome (MDS). NGS can be used for various applications: (i) in the diagnostic process to discriminate between MDS and other diseases such as aplastic anaemia, myeloproliferative disorders and idiopathic cytopenias; (ii) for classification, for example, where the presence of SF3B1 mutation is one criterion for the ring sideroblast anaemia subgroups in the World Health Organization 2016 classification; (iii) for identification of patients suitable for targeted therapy (e.g. IDH1/2 inhibitors); (iv) for prognostication, for example, where specific mutations (e.g. TP53 and RUNX1) are associated with inferior prognosis, whereas others (e.g. SF3B1) are associated with superior prognosis; and (v) to monitor patients for progression or treatment failure. Most commonly, targeted sequencing for genes (normally 50-100 genes) reported to be recurrently mutated in myeloid disease is used. At present, NGS is rarely incorporated into clinical guidelines although an increasing number of studies have demonstrated the benefit of using NGS in the clinical management of MDS patients.
Collapse
Affiliation(s)
- M Tobiasson
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden.,Institution of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
| | - A O Kittang
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Section for Hematology, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
4
|
Gao C, Wang J, Li Y, Zhao H, Li R, Hou L, Zhang Y, Tian S, Liang H, Wang C, Chen X, Wang J. Incidence and risk of hematologic toxicities with hypomethylating agents in the treatment of myelodysplastic syndromes and acute myeloid leukopenia: A systematic review and meta-analysis. Medicine (Baltimore) 2018; 97:e11860. [PMID: 30142779 PMCID: PMC6112947 DOI: 10.1097/md.0000000000011860] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Hypomethylating agents (HMAs) are believed to have reliable efficacy in treating myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Meanwhile, the adverse events of HMAs have become an increasing concern. There is, however, no systematic meta-analysis available to evaluate overall hematologic toxicities for HMAs. In this meta-analysis, we aim to determine the risk of hematologic toxicities in patients treated with HMAs. METHODS Relevant studies were identified from PubMed, Embase, Cochrane Library, and the Clinical Trials. gov databases incepted to February 2018. All phase II and III trials meeting the inclusion criteria included adequate safety data. We calculated the relative risk (RR) of high-grade hematologic toxicities (HTEs) with corresponding 95% CI using Review Manager. The incidences of HTEs were also evaluated by R. Heterogeneity was calculated and reported mainly via I analyses. RESULTS A total of 2337 MDS or AML patients from 14 studies were identified in this meta-analysis. The overall incidences of high-grade hematologic toxicities in patients who received HMAs were: 27% of the patients with anemia, 45% with neutropenia, 38% with thrombocytopenia, and 25% with febrile neutropenia, respectively. There was a significantly increased RR of neutropenia and thrombocytopenia using HMAs, in comparison with conventional care regimens (CCR) based on the drug type (decitabine vs azacitidine). CONCLUSIONS We conclude that the use of HMAs are associated with an increased risk of neutropenia and thrombocytopenia in MDS or AML patients, and our results also demonstrate that HMAs exposure does not significantly increase the risk of high-grade anemia, leukopenia, or febrile neutropenia compared with CCR.
Collapse
Affiliation(s)
- Chong Gao
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Dongcheng District
| | - Jia Wang
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Dongcheng District
| | - Ya Li
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Dongcheng District
| | - Huan Zhao
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Dongcheng District
| | - Ruibai Li
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Dongcheng District
| | - Li Hou
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Dongcheng District
| | - Yayue Zhang
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Dongcheng District
| | - Shaodan Tian
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Dongcheng District
| | - Huan Liang
- Cross Slope Community Health Service Station, Dongcheng District Community Health Service Management Center, Beijing, China
| | - Chong Wang
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Dongcheng District
| | - Xinyi Chen
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Dongcheng District
| | - Jing Wang
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Dongcheng District
| |
Collapse
|
5
|
Tobiasson M, Abdulkadir H, Lennartsson A, Katayama S, Marabita F, De Paepe A, Karimi M, Krjutskov K, Einarsdottir E, Grövdal M, Jansson M, Ben Azenkoud A, Corddedu L, Lehmann S, Ekwall K, Kere J, Hellström-Lindberg E, Ungerstedt J. Comprehensive mapping of the effects of azacitidine on DNA methylation, repressive/permissive histone marks and gene expression in primary cells from patients with MDS and MDS-related disease. Oncotarget 2018; 8:28812-28825. [PMID: 28427179 PMCID: PMC5438694 DOI: 10.18632/oncotarget.15807] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 02/01/2017] [Indexed: 02/07/2023] Open
Abstract
Azacitidine (Aza) is first-line treatment for patients with high-risk myelodysplastic syndromes (MDS), although its precise mechanism of action is unknown. We performed the first study to globally evaluate the epigenetic effects of Aza on MDS bone marrow progenitor cells assessing gene expression (RNA seq), DNA methylation (Illumina 450k) and the histone modifications H3K18ac and H3K9me3 (ChIP seq). Aza induced a general increase in gene expression with 924 significantly upregulated genes but this increase showed no correlation with changes in DNA methylation or H3K18ac, and only a weak association with changes in H3K9me3. Interestingly, we observed activation of transcripts containing 15 endogenous retroviruses (ERVs) confirming previous cell line studies. DNA methylation decreased moderately in 99% of all genes, with a median β-value reduction of 0.018; the most pronounced effects seen in heterochromatin. Aza-induced hypomethylation correlated significantly with change in H3K9me3. The pattern of H3K18ac and H3K9me3 displayed large differences between patients and healthy controls without any consistent pattern induced by Aza. We conclude that the marked induction of gene expression only partly could be explained by epigenetic changes, and propose that activation of ERVs may contribute to the clinical effects of Aza in MDS.
Collapse
Affiliation(s)
- Magnus Tobiasson
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Division of Hematology Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Hani Abdulkadir
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Division of Hematology Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Andreas Lennartsson
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm County, Sweden
| | - Shintaro Katayama
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm County, Sweden
| | - Francesco Marabita
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden.,National Bioinformatics Infrastructure Sweden, Stockholm, Sweden
| | - Ayla De Paepe
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Division of Hematology Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Mohsen Karimi
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Division of Hematology Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Kaarel Krjutskov
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm County, Sweden.,Molecular Neurology Research Program, University of Helsinki, and Folkhälsan Institute of Genetics, Helsinki, Finland.,Competence Centre on Health Technologies, Tartu, Estonia
| | - Elisabet Einarsdottir
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm County, Sweden.,Molecular Neurology Research Program, University of Helsinki, and Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Michael Grövdal
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Division of Hematology Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Monika Jansson
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Division of Hematology Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Asmaa Ben Azenkoud
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Division of Hematology Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Lina Corddedu
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm County, Sweden
| | - Sören Lehmann
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Division of Hematology Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden.,Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Karl Ekwall
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm County, Sweden
| | - Juha Kere
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm County, Sweden.,Molecular Neurology Research Program, University of Helsinki, and Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Eva Hellström-Lindberg
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Division of Hematology Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Johanna Ungerstedt
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Division of Hematology Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| |
Collapse
|
6
|
DNA Methylation Events as Markers for Diagnosis and Management of Acute Myeloid Leukemia and Myelodysplastic Syndrome. DISEASE MARKERS 2017; 2017:5472893. [PMID: 29038614 PMCID: PMC5606093 DOI: 10.1155/2017/5472893] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/17/2017] [Accepted: 07/30/2017] [Indexed: 01/18/2023]
Abstract
During the onset and progression of hematological malignancies, many changes occur in cellular epigenome, such as hypo- or hypermethylation of CpG islands in promoter regions. DNA methylation is an epigenetic modification that regulates gene expression and is a key event for tumorigenesis. The continuous search for biomarkers that signal early disease, indicate prognosis, and act as therapeutic targets has led to studies investigating the role of DNA in cancer onset and progression. This review focuses on DNA methylation changes as potential biomarkers for diagnosis, prognosis, response to treatment, and early toxicity in acute myeloid leukemia and myelodysplastic syndrome. Here, we report that distinct changes in DNA methylation may alter gene function and drive malignant cellular transformation during several stages of leukemogenesis. Most of these modifications occur at an early stage of disease and may predict myeloid/lymphoid transformation or response to therapy, which justifies its use as a biomarker for disease onset and progression. Methylation patterns, or its dynamic change during treatment, may also be used as markers for patient stratification, disease prognosis, and response to treatment. Further investigations of methylation modifications as therapeutic biomarkers, which may correlate with therapeutic response and/or predict treatment toxicity, are still warranted.
Collapse
|
7
|
Wang H, Zhang TT, Jin S, Liu H, Zhang X, Ruan CG, Wu DP, Han Y, Wang XQ. Pyrosequencing quantified methylation level of miR-124 predicts shorter survival for patients with myelodysplastic syndrome. Clin Epigenetics 2017; 9:91. [PMID: 28861128 PMCID: PMC5577794 DOI: 10.1186/s13148-017-0388-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 08/14/2017] [Indexed: 01/22/2023] Open
Abstract
Background Aberrant CpG island methylation has been increasingly recognized as a common event in myelodysplastic syndrome (MDS). To date, most of the previous studies of miR-124 in MDS have focused on epigenetic changes and little is known about the underlying mechanism through which miR-124 regulates CDK6 expression. Results In the present study, we employed pyrosequencing analysis to quantify the methylation levels of upstream regions of the miR-124 genes (miR-124-1, miR-124-2 and miR-124-3) in 56 primary MDS patients. We found the three miR-124 genes were methylated in MDS patients. Univariate analysis revealed that the World Health Organization (WHO) classification, marrow blast count, karyotype, International Prognostic Scoring System (IPSS), mean corpuscular volume, as well as high methylation of miR-124-1, miR-124-2 and miR-124-3 were significantly related to overall survival. In leukaemia-free survival, patients who were older and had an advanced WHO classification, high marrow blast counts, high IPSS risk and high methylation of miR-124-1 and miR-124-2 progressed rapidly to acute myeloid leukaemia. Multivariate analysis demonstrated that high methylation of miR-124-3 was an independent factor of overall survival. Median survival of patients with high miR-124-3 methylation was significantly shorter (7.6 months) than patients with low methylation (32.7 months; P = 0.010). A functional study revealed that silencing of miR-124 resulted in upregulation of its target gene, cyclin dependent kinase CDK6, which in turn promoted cell proliferation in the MDS cell line SKM-1. Conclusions High methylation of miR-124-3 predicts shorter survival for patients with MDS, which may be a useful prognostic marker in MDS. Electronic supplementary material The online version of this article (doi:10.1186/s13148-017-0388-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hong Wang
- Jiangsu Institute of Haematology, Institute of Blood and Marrow Transplantation, Department of Haematology, Collaborative Innovation of Haematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, 188 Shi Zi Street, Suzhou, 215000 China
| | - Tong-Tong Zhang
- Jiangsu Institute of Haematology, Institute of Blood and Marrow Transplantation, Department of Haematology, Collaborative Innovation of Haematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, 188 Shi Zi Street, Suzhou, 215000 China
| | - Song Jin
- Jiangsu Institute of Haematology, Institute of Blood and Marrow Transplantation, Department of Haematology, Collaborative Innovation of Haematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, 188 Shi Zi Street, Suzhou, 215000 China
| | - Hong Liu
- Jiangsu Institute of Haematology, Institute of Blood and Marrow Transplantation, Department of Haematology, Collaborative Innovation of Haematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, 188 Shi Zi Street, Suzhou, 215000 China
| | - Xiang Zhang
- Jiangsu Institute of Haematology, Institute of Blood and Marrow Transplantation, Department of Haematology, Collaborative Innovation of Haematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, 188 Shi Zi Street, Suzhou, 215000 China
| | - Chang-Geng Ruan
- Jiangsu Institute of Haematology, Institute of Blood and Marrow Transplantation, Department of Haematology, Collaborative Innovation of Haematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, 188 Shi Zi Street, Suzhou, 215000 China
| | - De-Pei Wu
- Jiangsu Institute of Haematology, Institute of Blood and Marrow Transplantation, Department of Haematology, Collaborative Innovation of Haematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, 188 Shi Zi Street, Suzhou, 215000 China
| | - Yue Han
- Jiangsu Institute of Haematology, Institute of Blood and Marrow Transplantation, Department of Haematology, Collaborative Innovation of Haematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, 188 Shi Zi Street, Suzhou, 215000 China
| | - Xiao-Qin Wang
- Department of Haematology, Huashan Hospital of Fudan University, 12 Wulumuqi Road Central, Shanghai, 200040 China
| |
Collapse
|
8
|
Luskin MR, Stone RM. Can Minimal Residual Disease Determination in Acute Myeloid Leukemia Be Used in Clinical Practice? J Oncol Pract 2017; 13:471-480. [DOI: 10.1200/jop.2017.021675] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In acute myeloid leukemia (AML) that is in complete remission, minimal residual disease (MRD) is presumed to be present, though not morphologically evident. Advances in diagnostics now permit the detection and quantification of MRD in AML by several techniques. The level of MRD after induction and consolidation therapy correlates with disease sensitivity to chemotherapy and has greater power to predict long-term survival than patient and disease characteristics that are available at diagnosis, including genetic information. A unique advantage of MRD is that it is an integrated measure of the impact and interaction of genetics, epigenetics, host immune milieu, bone marrow environment, and drug sensitivity on disease response to treatment. Here, we review the main techniques for MRD assessment in AML, including polymerase chain reaction, multiparameter flow cytometry, and next-generation sequencing, with a focus on method-specific and general limitations to the optimal employment of MRD techniques for the determination of AML prognosis. We also review the data that establish the prognostic and predictive value of MRD assessment in AML. Finally, we provide recommendations for the use of MRD in the care of patients with AML in clinical practice today, including whether it should influence treatment decisions.
Collapse
Affiliation(s)
- Marlise R. Luskin
- Dana-Farber Cancer Institute; and Harvard Medical School, Boston, MA
| | - Richard M. Stone
- Dana-Farber Cancer Institute; and Harvard Medical School, Boston, MA
| |
Collapse
|
9
|
Li Y, Xu Q, Lv N, Wang L, Zhao H, Wang X, Guo J, Chen C, Li Y, Yu L. Clinical implications of genome-wide DNA methylation studies in acute myeloid leukemia. J Hematol Oncol 2017; 10:41. [PMID: 28153026 PMCID: PMC5290606 DOI: 10.1186/s13045-017-0409-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 01/27/2017] [Indexed: 01/01/2023] Open
Abstract
Acute myeloid leukemia (AML) is the most common type of acute leukemia in adults. AML is a heterogeneous malignancy characterized by distinct genetic and epigenetic abnormalities. Recent genome-wide DNA methylation studies have highlighted an important role of dysregulated methylation signature in AML from biological and clinical standpoint. In this review, we will outline the recent advances in the methylome study of AML and overview the impacts of DNA methylation on AML diagnosis, treatment, and prognosis.
Collapse
Affiliation(s)
- Yan Li
- Department of Hematology and BMT center, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.,Department of Hematology, Hainan Branch of Chinese PLA General Hospital, Sanya, 572013, Hainan Province, China
| | - Qingyu Xu
- Department of Hematology and BMT center, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.,Medical school of Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Na Lv
- Department of Hematology and BMT center, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Lili Wang
- Department of Hematology and BMT center, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Hongmei Zhao
- Annoroad Gene Technology Co. Ltd, Beijing, 100176, China
| | - Xiuli Wang
- Annoroad Gene Technology Co. Ltd, Beijing, 100176, China
| | - Jing Guo
- Annoroad Gene Technology Co. Ltd, Beijing, 100176, China
| | - Chongjian Chen
- Annoroad Gene Technology Co. Ltd, Beijing, 100176, China
| | - Yonghui Li
- Department of Hematology and BMT center, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Li Yu
- Department of Hematology and BMT center, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| |
Collapse
|
10
|
Kostroma II, Gritsaev SV, Sidorova ZY, Tiranova SA, Svitina SP, Drizhun YS, Chubukina ZV, Martynkevich IS, Kapustin SI, Bessmeltsev SS. [Aberrant methylation of the promoter regions of the SOX7 and p15INK4b genes and Wnt signaling pathway antagonists in patients with acute myeloid leukemias]. TERAPEVT ARKH 2017; 88:31-36. [PMID: 27459612 DOI: 10.17116/terarkh201688731-36] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
AIM to investigate the methylation status of the SOX7 and p15NK4b genes and Wnt signaling pathway antagonists in patients with acute myeloid leukemia (AML) in order to assess the association of the rate of aberrant methylation (AM) with the morphological variant and pattern of chromosomal aberrations, as well as the impact of the methylation status on survival. SUBJECTS AND METHODS The data of 57 AML patients aged 20 to 79 years were analyzed. The methylation status of the genes was studied by methylation-specific polymerase chain reaction. RESULTS The signs of the AM of ≥1 gene were detected in 52 (91.2%) of the 57 patients. The most common finding was AM of simultaneously 2 or 3 genes: in 29.8 and 21.1% of the patients, respectively. Concurrent methylation of 3-5 genes proved to be a more frequent finding in AML patients with myelodysplasia: in 7 (70%) of 10 patients. The proportion of patients with methylation of 5 genes was considerably higher in a group of patients with a complex karyotype: 50% versus 8.3% among other patients (odds ratio: 11.0; 95% confidence interval 2.0 to 61.6; p=0.01). There were no differences in the median overall and relapse-free survival rates in patients with a normal karyotype and without FLT3 and NPM mutations, who received induction therapy, in relation to the number of genes with AM. CONCLUSION AM of the p15NK4b and SOX7 genes and Wnt signaling pathway antagonists is detected in the majority of patients with AML, which allows hypomethylating agents to be recommended for the treatment of patients who cannot use intensive cytostatic therapy for different reasons. The detection of a large number of genes with the aberrant methylation status in most AML patients with myelodysplasia or a complex karyotype serves as the basis for initiating trials to evaluate the efficiency of a combination of 5-azacytidine and cytostatics.
Collapse
Affiliation(s)
- I I Kostroma
- Russian Research Institute of Hematology and Transfusiology, Federal Biomedical Agency of Russia, Moscow, Russia
| | - S V Gritsaev
- Russian Research Institute of Hematology and Transfusiology, Federal Biomedical Agency of Russia, Moscow, Russia
| | - Zh Yu Sidorova
- Russian Research Institute of Hematology and Transfusiology, Federal Biomedical Agency of Russia, Moscow, Russia
| | - S A Tiranova
- Russian Research Institute of Hematology and Transfusiology, Federal Biomedical Agency of Russia, Moscow, Russia
| | - S P Svitina
- Russian Research Institute of Hematology and Transfusiology, Federal Biomedical Agency of Russia, Moscow, Russia
| | - Yu S Drizhun
- Russian Research Institute of Hematology and Transfusiology, Federal Biomedical Agency of Russia, Moscow, Russia
| | - Zh V Chubukina
- Russian Research Institute of Hematology and Transfusiology, Federal Biomedical Agency of Russia, Moscow, Russia
| | - I S Martynkevich
- Russian Research Institute of Hematology and Transfusiology, Federal Biomedical Agency of Russia, Moscow, Russia
| | - S I Kapustin
- Russian Research Institute of Hematology and Transfusiology, Federal Biomedical Agency of Russia, Moscow, Russia
| | - S S Bessmeltsev
- Russian Research Institute of Hematology and Transfusiology, Federal Biomedical Agency of Russia, Moscow, Russia
| |
Collapse
|
11
|
Luskin MR, Gimotty PA, Smith C, Loren AW, Figueroa ME, Harrison J, Sun Z, Tallman MS, Paietta EM, Litzow MR, Melnick AM, Levine RL, Fernandez HF, Luger SM, Carroll M, Master SR, Wertheim GBW. A clinical measure of DNA methylation predicts outcome in de novo acute myeloid leukemia. JCI Insight 2016; 1. [PMID: 27446991 DOI: 10.1172/jci.insight.87323] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Variable response to chemotherapy in acute myeloid leukemia (AML) represents a major treatment challenge. Clinical and genetic features incompletely predict outcome. The value of clinical epigenetic assays for risk classification has not been extensively explored. We assess the prognostic implications of a clinical assay for multilocus DNA methylation on adult patients with de novo AML. METHODS We performed multilocus DNA methylation assessment using xMELP on samples and calculated a methylation statistic (M-score) for 166 patients from UPENN with de novo AML who received induction chemotherapy. The association of M-score with complete remission (CR) and overall survival (OS) was evaluated. The optimal M-score cut-point for identifying groups with differing survival was used to define a binary M-score classifier. This classifier was validated in an independent cohort of 383 patients from the Eastern Cooperative Oncology Group Trial 1900 (E1900; NCT00049517). RESULTS A higher mean M-score was associated with death and failure to achieve CR. Multivariable analysis confirmed that a higher M-score was associated with death (P = 0.011) and failure to achieve CR (P = 0.034). Median survival was 26.6 months versus 10.6 months for low and high M-score groups. The ability of the M-score to perform as a classifier was confirmed in patients ≤ 60 years with intermediate cytogenetics and patients who achieved CR, as well as in the E1900 validation cohort. CONCLUSION The M-score represents a valid binary prognostic classifier for patients with de novo AML. The xMELP assay and associated M-score can be used for prognosis and should be further investigated for clinical decision making in AML patients.
Collapse
Affiliation(s)
- Marlise R Luskin
- Division of Hematology and Oncology, Abramson Cancer Center at the University of Pennsylvania (UPENN), Philadelphia, Pennsylvania, USA
| | - Phyllis A Gimotty
- Department of Biostatistics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania (UPENN), Philadelphia, Pennsylvania, USA
| | - Catherine Smith
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Alison W Loren
- Division of Hematology and Oncology, Abramson Cancer Center at the University of Pennsylvania (UPENN), Philadelphia, Pennsylvania, USA
| | | | - Jenna Harrison
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Zhuoxin Sun
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | | | | | - Ari M Melnick
- Hematology and Oncology Division, Weill Cornell Medical College, New York, New York, USA
| | - Ross L Levine
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Selina M Luger
- Division of Hematology and Oncology, Abramson Cancer Center at the University of Pennsylvania (UPENN), Philadelphia, Pennsylvania, USA
| | - Martin Carroll
- Division of Hematology and Oncology, Abramson Cancer Center at the University of Pennsylvania (UPENN), Philadelphia, Pennsylvania, USA; Philadelphia Veterans Administration Medical Center, Philadelphia, Pennsylvania, USA
| | - Stephen R Master
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Gerald B W Wertheim
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
12
|
Laille E, Shi T, Garcia-Manero G, Cogle CR, Gore SD, Hetzer J, Kumar K, Skikne B, MacBeth KJ. Pharmacokinetics and Pharmacodynamics with Extended Dosing of CC-486 in Patients with Hematologic Malignancies. PLoS One 2015; 10:e0135520. [PMID: 26296092 PMCID: PMC4546409 DOI: 10.1371/journal.pone.0135520] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 07/20/2015] [Indexed: 12/31/2022] Open
Abstract
CC-486 (oral azacitidine) is an epigenetic modifier in development for patients with myelodysplastic syndromes and acute myeloid leukemia. In part 1 of this two-part study, a 7-day CC-486 dosing schedule showed clinical activity, was generally well tolerated, and reduced DNA methylation. Extending dosing of CC-486 beyond 7 days would increase duration of azacitidine exposure. We hypothesized that extended dosing would therefore provide more sustained epigenetic activity. Reported here are the pharmacokinetic (PK) and pharmacodynamic (PD) profiles of CC-486 extended dosing schedules in patients with myelodysplastic syndromes (MDS), chronic myelomonocytic leukemia (CMML) or acute myeloid leukemia (AML) from part 2 of this study. PK and/or PD data were available for 59 patients who were sequentially assigned to 1 of 4 extended CC-486 dosing schedules: 300mg once-daily or 200mg twice-daily for 14 or 21 days per 28-day cycle. Both 300mg once-daily schedules and the 200mg twice-daily 21-day schedule significantly (all P < .05) reduced global DNA methylation in whole blood at all measured time points (days 15, 22, and 28 of the treatment cycle), with sustained hypomethylation at cycle end compared with baseline. CC-486 exposures and reduced DNA methylation were significantly correlated. Patients who had a hematologic response had significantly greater methylation reductions than non-responding patients. These data demonstrate that extended dosing of CC-486 sustains epigenetic effects through the treatment cycle.
Collapse
MESH Headings
- Administration, Oral
- Aged
- Aged, 80 and over
- Antimetabolites, Antineoplastic/blood
- Antimetabolites, Antineoplastic/pharmacokinetics
- Antimetabolites, Antineoplastic/therapeutic use
- Area Under Curve
- Azacitidine/blood
- Azacitidine/pharmacokinetics
- Azacitidine/therapeutic use
- DNA Methylation
- Dose-Response Relationship, Drug
- Drug Administration Schedule
- Epigenesis, Genetic
- Half-Life
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myelomonocytic, Chronic/drug therapy
- Leukemia, Myelomonocytic, Chronic/genetics
- Leukemia, Myelomonocytic, Chronic/mortality
- Leukemia, Myelomonocytic, Chronic/pathology
- Male
- Middle Aged
- Myelodysplastic Syndromes/drug therapy
- Myelodysplastic Syndromes/genetics
- Myelodysplastic Syndromes/mortality
- Myelodysplastic Syndromes/pathology
- Survival Analysis
Collapse
Affiliation(s)
- Eric Laille
- Celgene Corporation, Summit, New Jersey, United States of America
- * E-mail:
| | - Tao Shi
- Celgene Corporation, Summit, New Jersey, United States of America
| | - Guillermo Garcia-Manero
- Department of Leukemia, University of Texas, MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Christopher R. Cogle
- Division of Hematology/Oncology, University of Florida, Gainesville, Florida, United States of America
| | - Steven D. Gore
- Yale Cancer Center, New Haven, Connecticut, United States of America
| | - Joel Hetzer
- Celgene Corporation, Summit, New Jersey, United States of America
| | - Keshava Kumar
- Celgene Corporation, Summit, New Jersey, United States of America
| | - Barry Skikne
- Celgene Corporation, Summit, New Jersey, United States of America
| | - Kyle J. MacBeth
- Celgene Corporation, Summit, New Jersey, United States of America
| |
Collapse
|
13
|
Wong JJL, Lau KA, Pinello N, Rasko JEJ. Epigenetic modifications of splicing factor genes in myelodysplastic syndromes and acute myeloid leukemia. Cancer Sci 2014; 105:1457-63. [PMID: 25220401 PMCID: PMC4462368 DOI: 10.1111/cas.12532] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 08/28/2014] [Accepted: 09/01/2014] [Indexed: 12/19/2022] Open
Abstract
Somatic mutations in splicing factor genes have frequently been reported in myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Although aberrant epigenetic changes are frequently implicated in blood cancers, their direct role in suppressing one or both alleles of critical splicing factors has not been previously examined. Here, we examined promoter DNA hypermethylation of nine splicing factors, SF3B1, SRSF2, U2AF1, ZRSR2, SF3A1, HNRNPR, MATR3, ZFR, and YBX3 in 10 leukemic cell lines and 94 MDS or AML patient samples from the Australasian Leukemia and Lymphoma Group Tissue Bank. The only evidence of epigenetic effects was hypermethylation of the YBX3 promoter in U937 cells in conjunction with an enrichment of histone marks associated with gene silencing. In silico analysis of DNA methylation data for 173 AML samples generated by the Cancer Genome Atlas Research Network revealed promoter hypermethylation of the gene encoding Y box binding protein 3, YBX3, in 11/173 (6.4%) AML cases, which was significantly associated with reduced mRNA expression (P < 0.0001). Hypermethylation of the ZRSR2 promoter was also detected in 7/173 (4%) cases but was not associated with decreased mRNA expression (P = 0.1204). Hypermethylation was absent at the promoter of seven other splicing factor genes in all cell lines and patient samples examined. We conclude that DNA hypermethylation does not frequently silence splicing factors in MDS and AML. However, in the case of YBX3, promoter hypermethylation-induced downregulation may contribute to the pathogenesis or maintenance of AML.
Collapse
Affiliation(s)
- Justin J-L Wong
- Gene and Stem Cell Therapy Program, Centenary Institute, Camperdown, New South Wales, Australia; Sydney Medical School, University of Sydney, Camperdown, New South Wales, Australia
| | | | | | | |
Collapse
|
14
|
Novel drugs for older patients with acute myeloid leukemia. Leukemia 2014; 29:760-9. [PMID: 25142817 DOI: 10.1038/leu.2014.244] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 07/30/2014] [Accepted: 08/04/2014] [Indexed: 02/07/2023]
Abstract
Acute myeloid leukemia (AML) is the second most common form of leukemia and the most frequent cause of leukemia-related deaths in the United States. The incidence of AML increases with advancing age and the prognosis for patients with AML worsens substantially with increasing age. Many older patients are ineligible for intensive treatment and require other therapeutic approaches to optimize clinical outcome. To address this treatment gap, novel agents with varying mechanisms of action targeting different cellular processes are currently in development. Hypomethylating agents (azacitidine, decitabine, SGI-110), histone deacetylase inhibitors (vorinostat, pracinostat, panobinostat), FMS-like tyrosine kinase receptor-3 inhibitors (quizartinib, sorafenib, midostaurin, crenolanib), cytotoxic agents (clofarabine, sapacitabine, vosaroxin), cell cycle inhibitors (barasertib, volasertib, rigosertib) and monoclonal antibodies (gentuzumab ozogamicin, lintuzumab-Ac225) represent some of these promising new treatments. This review provides an overview of novel agents that have either completed or are currently in ongoing phase III trials in patients with previously untreated AML for whom intensive treatment is not an option. Other potential drugs in earlier stages of development will also be addressed in this review.
Collapse
|
15
|
Tobiasson M, Dybedahl I, Holm MS, Karimi M, Brandefors L, Garelius H, Grövdal M, Högh-Dufva I, Grønbæk K, Jansson M, Marcher C, Nilsson L, Kittang AO, Porwit A, Saft L, Möllgård L, Hellström-Lindberg E. Limited clinical efficacy of azacitidine in transfusion-dependent, growth factor-resistant, low- and Int-1-risk MDS: Results from the nordic NMDSG08A phase II trial. Blood Cancer J 2014; 4:e189. [PMID: 24608733 PMCID: PMC3972706 DOI: 10.1038/bcj.2014.8] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 01/24/2014] [Accepted: 01/29/2014] [Indexed: 02/08/2023] Open
Abstract
This prospective phase II study evaluated the efficacy of azacitidine (Aza)+erythropoietin (Epo) in transfusion-dependent patients with lower-risk myelodysplastic syndrome (MDS). Patients ineligible for or refractory to full-dose Epo+granulocyte colony stimulation factors for >8 weeks and a transfusion need of 4 units over 8 weeks were included. Aza 75 mg m(-2) d(-1), 5/28 days, was given for six cycles; non-responding patients received another three cycles combined with Epo 60 000 units per week. Primary end point was transfusion independence (TI). All patients underwent targeted mutational screen for 42 candidate genes. Thirty enrolled patients received one cycle of Aza. Ten patients discontinued the study early, 7 due to adverse events including 2 deaths. Thirty-eight serious adverse events were reported, the most common being infection. Five patients achieved TI after six cycles and one after Aza+Epo, giving a total response rate of 20%. Mutational screening revealed a high frequency of recurrent mutations. Although no single mutation predicted for response, SF3A1 (n=3) and DNMT3A (n=4) were only observed in non-responders. We conclude that Aza can induce TI in severely anemic MDS patients, but efficacy is limited, toxicity substantial and most responses of short duration. This treatment cannot be generally recommended in lower-risk MDS. Mutational screening revealed a high frequency of mutations.
Collapse
Affiliation(s)
- M Tobiasson
- Division of Hematology, Department of Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - I Dybedahl
- Department of Hematology, Rikshospitalet, Oslo, Norway
| | - M S Holm
- Department of Hematology, Århus University Hospital, Århus, Denmark
| | - M Karimi
- Karolinska Institutet, Institution for Medicine, Stockholm, Sweden
| | - L Brandefors
- Department of Medicine, Sunderbyn Hospital, Luleå, Sweden
| | - H Garelius
- Department of Hematology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - M Grövdal
- Department of Medicine, SöDersjukhuset, Stockholm, Sweden
| | - I Högh-Dufva
- Department of Hematology, Herlev University Hospital, Copenhagen, Denmark
| | - K Grønbæk
- Department of Hematology, Rigshospitalet University Hospital, Copenhagen, Denmark
| | - M Jansson
- Karolinska Institutet, Institution for Medicine, Stockholm, Sweden
| | - C Marcher
- Department of Hematology, Odense University Hospital, Odense, Denmark
| | - L Nilsson
- Department of Hematology, Lund University Hospital, Lund, Sweden
| | - A O Kittang
- Department of Medicine and University of Bergen, K2 Clinical Institute 2, Haukeland University Hospital, Bergen, Norway
| | - A Porwit
- Division of Hematology, Department of Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - L Saft
- Division of Hematology, Department of Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - L Möllgård
- Division of Hematology, Department of Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - E Hellström-Lindberg
- Division of Hematology, Department of Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
16
|
Pellagatti A, Benner A, Mills KI, Cazzola M, Giagounidis A, Perry J, Malcovati L, Della Porta MG, Jädersten M, Verma A, McDonald EJ, Killick S, Hellström-Lindberg E, Bullinger L, Wainscoat JS, Boultwood J. Identification of gene expression-based prognostic markers in the hematopoietic stem cells of patients with myelodysplastic syndromes. J Clin Oncol 2013; 31:3557-64. [PMID: 24002510 DOI: 10.1200/jco.2012.45.5626] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
PURPOSE The diagnosis of patients with myelodysplastic syndromes (MDS) is largely dependent on morphologic examination of bone marrow aspirates. Several criteria that form the basis of the classifications and scoring systems most commonly used in clinical practice are affected by operator-dependent variation. To identify standardized molecular markers that would allow prediction of prognosis, we have used gene expression profiling (GEP) data on CD34+ cells from patients with MDS to determine the relationship between gene expression levels and prognosis. PATIENTS AND METHODS GEP data on CD34+ cells from 125 patients with MDS with a minimum 12-month follow-up since date of bone marrow sample collection were included in this study. Supervised principal components and lasso penalized Cox proportional hazards regression (Coxnet) were used for the analysis. RESULTS We identified several genes, the expression of which was significantly associated with survival of patients with MDS, including LEF1, CDH1, WT1, and MN1. The Coxnet predictor, based on expression data on 20 genes, outperformed other predictors, including one that additionally used clinical information. Our Coxnet gene signature based on CD34+ cells significantly identified a separation of patients with good or bad prognosis in an independent GEP data set based on unsorted bone marrow mononuclear cells, demonstrating that our signature is robust and may be applicable to bone marrow cells without the need to isolate CD34+ cells. CONCLUSION We present a new, valuable GEP-based signature for assessing prognosis in MDS. GEP-based signatures correlating with clinical outcome may significantly contribute to a refined risk classification of MDS.
Collapse
Affiliation(s)
- Andrea Pellagatti
- Andrea Pellagatti, Janet Perry, James S. Wainscoat, and Jacqueline Boultwood, University of Oxford, Oxford; Ken I. Mills, Queen's University Belfast, Belfast; Emma-Jane McDonald and Sally Killick, Royal Bournemouth Hospital, Bournemouth, United Kingdom; Axel Benner, German Cancer Research Center, Heidelberg; Aristoteles Giagounidis, St Johannes Hospital, Duisburg; Lars Bullinger, University Hospital of Ulm, Ulm, Germany; Mario Cazzola, Luca Malcovati, and Matteo G. Della Porta, Fondazione Istituto di Ricovera e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Martin Jädersten and Eva Hellström-Lindberg, Karolinska Institutet, Stockholm, Sweden; and Amit Verma, Albert Einstein College of Medicine, New York, NY
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Poloni A, Goteri G, Zizzi A, Serrani F, Trappolini S, Costantini B, Mariani M, Olivieri A, Catarini M, Centurioni R, Alesiani F, Giantomassi F, Stramazzotti D, Biagetti S, Alfonsi S, Berardinelli E, Leoni P. Prognostic role of immunohistochemical analysis of 5 mc in myelodysplastic syndromes. Eur J Haematol 2013; 91:219-227. [DOI: 10.1111/ejh.12145] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2013] [Indexed: 12/19/2022]
Affiliation(s)
- Antonella Poloni
- Clinica di Ematologia; Dipartimento di Scienze Cliniche e Molecolari; Università Politecnica Marche; Ancona; Italy
| | - Gaia Goteri
- Anatomia Patologica; Dipartimento di Neuroscienze; Università Politecnica Marche; Ancona; Italy
| | - Antonio Zizzi
- Anatomia Patologica; Dipartimento di Neuroscienze; Università Politecnica Marche; Ancona; Italy
| | - Federica Serrani
- Clinica di Ematologia; Dipartimento di Scienze Cliniche e Molecolari; Università Politecnica Marche; Ancona; Italy
| | - Silvia Trappolini
- Clinica di Ematologia; Dipartimento di Scienze Cliniche e Molecolari; Università Politecnica Marche; Ancona; Italy
| | - Benedetta Costantini
- Clinica di Ematologia; Dipartimento di Scienze Cliniche e Molecolari; Università Politecnica Marche; Ancona; Italy
| | - Marianna Mariani
- Clinica di Ematologia; Dipartimento di Scienze Cliniche e Molecolari; Università Politecnica Marche; Ancona; Italy
| | - Attilio Olivieri
- Clinica di Ematologia; Dipartimento di Scienze Cliniche e Molecolari; Università Politecnica Marche; Ancona; Italy
| | | | | | | | - Federica Giantomassi
- Anatomia Patologica; Dipartimento di Neuroscienze; Università Politecnica Marche; Ancona; Italy
| | - Daniela Stramazzotti
- Anatomia Patologica; Dipartimento di Neuroscienze; Università Politecnica Marche; Ancona; Italy
| | - Simona Biagetti
- Anatomia Patologica; Dipartimento di Neuroscienze; Università Politecnica Marche; Ancona; Italy
| | - Simona Alfonsi
- Anatomia Patologica; Dipartimento di Neuroscienze; Università Politecnica Marche; Ancona; Italy
| | - Eleonora Berardinelli
- Clinica di Ematologia; Dipartimento di Scienze Cliniche e Molecolari; Università Politecnica Marche; Ancona; Italy
| | - Pietro Leoni
- Clinica di Ematologia; Dipartimento di Scienze Cliniche e Molecolari; Università Politecnica Marche; Ancona; Italy
| |
Collapse
|
18
|
Yu J, Peng Y, Wu LC, Xie Z, Deng Y, Hughes T, He S, Mo X, Chiu M, Wang QE, He X, Liu S, Grever MR, Chan KK, Liu Z. Curcumin down-regulates DNA methyltransferase 1 and plays an anti-leukemic role in acute myeloid leukemia. PLoS One 2013; 8:e55934. [PMID: 23457487 PMCID: PMC3572185 DOI: 10.1371/journal.pone.0055934] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 01/08/2013] [Indexed: 12/21/2022] Open
Abstract
Bioactive components from dietary supplements such as curcumin may represent attractive agents for cancer prevention or treatment. DNA methylation plays a critical role in acute myeloid leukemia (AML) development, and presents an excellent target for treatment of this disease. However, it remains largely unknown how curcumin, a component of the popular Indian spice turmeric, plays a role in DNA hypomethylation to reactivate silenced tumor suppressor genes and to present a potential treatment option for AML. Here we show that curcumin down-regulates DNMT1 expression in AML cell lines, both in vitro and in vivo, and in primary AML cells ex vivo. Mechanistically, curcumin reduced the expression of positive regulators of DNMT1, p65 and Sp1, which correlated with a reduction in binding of these transcription factors to the DNMT1 promoter in AML cell lines. This curcumin-mediated down-regulation of DNMT1 expression was concomitant with p15INK4B tumor suppressor gene reactivation, hypomethylation of the p15INK4B promoter, G1 cell cycle arrest, and induction of tumor cell apoptosis in vitro. In mice implanted with the human AML MV4–11 cell line, administration of curcumin resulted in remarkable suppression of AML tumor growth. Collectively, our data indicate that curcumin shows promise as a potential treatment for AML, and our findings provide a basis for future studies to test the clinical efficacy of curcumin – whether used as a single agent or as an adjuvant – for AML treatment.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Caspases/metabolism
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Curcumin/pharmacology
- Curcumin/therapeutic use
- DNA (Cytosine-5-)-Methyltransferase 1
- DNA (Cytosine-5-)-Methyltransferases/genetics
- DNA (Cytosine-5-)-Methyltransferases/metabolism
- Down-Regulation/drug effects
- Female
- G1 Phase Cell Cycle Checkpoints/drug effects
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Mice
- Mice, Nude
- Promoter Regions, Genetic/drug effects
- Sp1 Transcription Factor/genetics
- Transcription Factor RelA/genetics
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Jianhua Yu
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail: (JY); (ZL)
| | - Yong Peng
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
| | - Lai-Chu Wu
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
- Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, Ohio, United States of America
| | - Zhiliang Xie
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States of America
| | - Youcai Deng
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Tiffany Hughes
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
| | - Shun He
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
| | - XiaoKui Mo
- Center for Biostatistics, The Ohio State University, Columbus, Ohio, United States of America
| | - Ming Chiu
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States of America
| | - Qi-En Wang
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
| | - Xiaoming He
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States of America
| | - Shujun Liu
- The Hormel Institute, University of Minnesota, Austin, Minnesota, United States of America
| | - Michael R. Grever
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
| | - Kenneth K. Chan
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States of America
| | - Zhongfa Liu
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail: (JY); (ZL)
| |
Collapse
|
19
|
Yang Y, Zhang Q, Xu F, Wu L, He Q, Li X. Tumor suppressor gene BLU is frequently downregulated by promoter hypermethylation in myelodysplastic syndrome. J Cancer Res Clin Oncol 2012; 138:729-37. [PMID: 22246278 DOI: 10.1007/s00432-012-1151-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2011] [Accepted: 01/02/2012] [Indexed: 01/16/2023]
Abstract
PURPOSE BLU methylation status was investigated in bone marrow mononuclear cells from newly diagnosed myelodysplastic syndrome (MDS) patients and patients who received 5-aza-2'-deoxycytidine (decitabine) treatment so as to determine the effect of BLU in the pathogenesis of MDS. METHODS Methylation-specific polymerase chain reaction and bisulfite sequencing were used to evaluate the methylation status of the promoter region of the BLU gene. BLU expression was investigated by using quantitative real-time reverse transcriptase polymerase chain reaction (qRT-PCR) and Western blot analysis. RESULTS Hypermethylation in the promoter region of BLU was detected in 34 of 79 (43%) newly diagnosed MDS patient samples and was significantly correlated with the loss of BLU mRNA and protein expression. There was a statistically significant difference in methylation frequency between the refractory anemia/refractory anemia with ringed sideroblasts/5q-syndrome (RA/RARS/5q-) group and the refractory anemia with excess blasts-1/-2 (RAEB-1/RAEB-2) group. A higher frequency of hypermethylation was observed in the intermediate-2/high-risk group compared to the low-risk/intermediate-1-risk group. The demethylating agent decitabine could partly reverse hypermethylation and restore the expression of the BLU gene. CONCLUSION BLU promoter hypermethylation frequently occurs in MDS cases, especially in higher risk MDS cases, and is significantly associated with the downregulated expression of BLU. BLU gene re-expression was induced in some MDS cases undergoing decitabine therapy. BLU may play a substantial role in the development and etiology of MDS.
Collapse
Affiliation(s)
- Yujuan Yang
- Department of Hematology, The Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | | | | | | | | | | |
Collapse
|
20
|
Wei Y, Gañán-Gómez I, Salazar-Dimicoli S, McCay SL, Garcia-Manero G. Histone methylation in myelodysplastic syndromes. Epigenomics 2012; 3:193-205. [PMID: 22122281 DOI: 10.2217/epi.11.9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Histone methylation is a type of epigenetic modification that is critical for the regulation of gene expression. Numerous studies have demonstrated that abnormalities of this newly characterized epigenetic modification are involved in the development of multiple diseases, including cancer. There is also emerging evidence for a link between histone methylation and the pathogenesis of myeloid neoplasms, including myelodysplastic syndromes (MDS). This article provides an overview of recent progress in the studies of histone methylation in myeloid malignancies, with an emphasis on MDS. We cover each type of histone methylation modification and their regulatory mechanisms, as well as their abnormalities in MDS or potential connections to MDS. We also summarize the recent progress in the development of inhibitors targeting histone methylation and their applications as potential therapeutic agents.
Collapse
Affiliation(s)
- Yue Wei
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | | | | | |
Collapse
|
21
|
Hypermethylation of SHP-1 promoter in patient with high-risk myelodysplastic syndrome and it predicts poor prognosis. Med Oncol 2012; 29:2359-63. [DOI: 10.1007/s12032-012-0163-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2011] [Accepted: 01/06/2012] [Indexed: 10/14/2022]
|
22
|
Abstract
Myeloid hematological malignancies are among the epigenetically best characterized neoplasms. The comparatively low number of recurring balanced and unbalanced chromosomal abnormalities as well as common genetic mutations has enabled scientists to relate epigenetic states to these. The ease of accessing malignant cells through bone marrow aspiration has certainly contributed to the fast expansion of knowledge. Even so, the clinical and pathogenetic relevance of epigenetic changes is still not known, and the field will certainly evolve very fast with the development of new analytic techniques. The first example of successful epigenetic therapy is seen in myeloid malignancies, in the high-risk myelodysplastic syndromes (MDS) which are routinely treated with the demethylating agent azacytidine.This chapter will concentrate on describing the epigenetic changes in acute myeloid leukemia (AML), chronic myeloid leukemia (CML) and MDS. An overview of clinical relevance and epigenetic therapeutic approaches is also made.
Collapse
Affiliation(s)
- Stefan Deneberg
- Center of Hematology, Karolinska University Hospital, Huddinge, Sweden.
| |
Collapse
|
23
|
Vigna E, Recchia AG, Madeo A, Gentile M, Bossio S, Mazzone C, Lucia E, Morabito L, Gigliotti V, Stefano LD, Caruso N, Servillo P, Franzese S, Fimognari F, Bisconte MG, Gentile C, Morabito F. Epigenetic regulation in myelodysplastic syndromes: implications for therapy. Expert Opin Investig Drugs 2011; 20:465-93. [DOI: 10.1517/13543784.2011.559164] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Ernesto Vigna
- Unità Operativa Complessa di Ematologia, Dipartimento Oncoematologico, Azienda Ospedaliera di Cosenza, Viale della Repubblica, 87100 Cosenza, Italy ;
| | - Anna Grazia Recchia
- Unità Operativa Complessa di Ematologia, Dipartimento Oncoematologico, Azienda Ospedaliera di Cosenza, Viale della Repubblica, 87100 Cosenza, Italy ;
| | - Antonio Madeo
- Unità Operativa Complessa di Ematologia, Dipartimento Oncoematologico, Azienda Ospedaliera di Cosenza, Viale della Repubblica, 87100 Cosenza, Italy ;
| | - Massimo Gentile
- Unità Operativa Complessa di Ematologia, Dipartimento Oncoematologico, Azienda Ospedaliera di Cosenza, Viale della Repubblica, 87100 Cosenza, Italy ;
| | - Sabrina Bossio
- Unità Operativa Complessa di Ematologia, Dipartimento Oncoematologico, Azienda Ospedaliera di Cosenza, Viale della Repubblica, 87100 Cosenza, Italy ;
| | - Carla Mazzone
- Unità Operativa Complessa di Ematologia, Dipartimento Oncoematologico, Azienda Ospedaliera di Cosenza, Viale della Repubblica, 87100 Cosenza, Italy ;
| | - Eugenio Lucia
- Unità Operativa Complessa di Ematologia, Dipartimento Oncoematologico, Azienda Ospedaliera di Cosenza, Viale della Repubblica, 87100 Cosenza, Italy ;
| | - Lucio Morabito
- Servicio de Hematología y Hemoterapia, Hospital Universitario de Canarias, La Laguna, Tenerife, Spain
| | - Vincenzo Gigliotti
- Unità Operativa Complessa di Ematologia, Dipartimento Oncoematologico, Azienda Ospedaliera di Cosenza, Viale della Repubblica, 87100 Cosenza, Italy ;
| | - Laura De Stefano
- Unità Operativa Complessa di Ematologia, Dipartimento Oncoematologico, Azienda Ospedaliera di Cosenza, Viale della Repubblica, 87100 Cosenza, Italy ;
| | - Nadia Caruso
- Unità Operativa Complessa di Ematologia, Dipartimento Oncoematologico, Azienda Ospedaliera di Cosenza, Viale della Repubblica, 87100 Cosenza, Italy ;
| | - Pasquale Servillo
- Unità Operativa Complessa di Ematologia, Dipartimento Oncoematologico, Azienda Ospedaliera di Cosenza, Viale della Repubblica, 87100 Cosenza, Italy ;
| | - Stefania Franzese
- Unità Operativa Complessa di Ematologia, Dipartimento Oncoematologico, Azienda Ospedaliera di Cosenza, Viale della Repubblica, 87100 Cosenza, Italy ;
| | | | - Maria Grazia Bisconte
- Unità Operativa Complessa di Ematologia, Dipartimento Oncoematologico, Azienda Ospedaliera di Cosenza, Viale della Repubblica, 87100 Cosenza, Italy ;
| | - Carlo Gentile
- Unità Operativa Complessa di Ematologia, Dipartimento Oncoematologico, Azienda Ospedaliera di Cosenza, Viale della Repubblica, 87100 Cosenza, Italy ;
| | - Fortunato Morabito
- Unità Operativa Complessa di Ematologia, Dipartimento Oncoematologico, Azienda Ospedaliera di Cosenza, Viale della Repubblica, 87100 Cosenza, Italy ;
| |
Collapse
|
24
|
Lin J, Wang YL, Qian J, Yao DM, Zhu ZH, Qian Z, Xu WR. Aberrant methylation of DNA-damage-inducible transcript 3 promoter is a common event in patients with myelodysplastic syndrome. Leuk Res 2010; 34:991-4. [DOI: 10.1016/j.leukres.2010.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Revised: 10/10/2009] [Accepted: 01/05/2010] [Indexed: 01/20/2023]
|
25
|
Grövdal M, Karimi M, Khan R, Aggerholm A, Antunovic P, Astermark J, Bernell P, Engström LM, Kjeldsen L, Linder O, Nilsson L, Olsson A, Holm MS, Tangen JM, Wallvik J, Oberg G, Hokland P, Jacobsen SE, Porwit A, Hellström-Lindberg E. Maintenance treatment with azacytidine for patients with high-risk myelodysplastic syndromes (MDS) or acute myeloid leukaemia following MDS in complete remission after induction chemotherapy. Br J Haematol 2010; 150:293-302. [PMID: 20497178 DOI: 10.1111/j.1365-2141.2010.08235.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
This prospective Phase II study is the first to assess the feasibility and efficacy of maintenance 5-azacytidine for older patients with high-risk myelodysplastic syndrome (MDS), chronic myelomonocytic leukaemia and MDS-acute myeloid leukaemia syndromes in complete remission (CR) after induction chemotherapy. Sixty patients were enrolled and treated by standard induction chemotherapy. Patients that reached CR started maintenance therapy with subcutaneous azacytidine, 5/28 d until relapse. Promoter-methylation status of CDKN2B (P15 ink4b), CDH1 and HIC1 was examined pre-induction, in CR and 6, 12 and 24 months post CR. Twenty-four (40%) patients achieved CR after induction chemotherapy and 23 started maintenance treatment with azacytidine. Median CR duration was 13.5 months, >24 months in 17% of the patients, and 18-30.5 months in the four patients with trisomy 8. CR duration was not associated with CDKN2B methylation status or karyotype. Median overall survival was 20 months. Hypermethylation of CDH1 was significantly associated with low CR rate, early relapse, and short overall survival (P = 0.003). 5-azacytidine treatment, at a dose of 60 mg/m(2) was well tolerated. Grade III-IV thrombocytopenia and neutropenia occurred after 9.5 and 30% of the cycles, respectively, while haemoglobin levels increased during treatment. 5-azacytidine treatment is safe, feasible and may be of benefit in a subset of patients.
Collapse
Affiliation(s)
- Michael Grövdal
- Division of Haematology, Department of Medicine, Centre for Experimental Haematology, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Epigenetic mechanisms, such as DNA methylation and histone modifications, drive stable, clonally propagated changes in gene expression and can therefore serve as molecular mediators of pathway dysfunction in neoplasia. Myelodysplastic syndrome (MDS) is characterized by frequent epigenetic abnormalities, including the hypermethylation of genes that control proliferation, adhesion, and other characteristic features of this leukemia. Aberrant DNA hypermethylation is associated with a poor prognosis in MDS that can be accounted for by more rapid progression to acute myeloid leukemia. In turn, treatment with drugs that modify epigenetic pathways (DNA methylation and histone deacetylation inhibitors) induces durable remissions and prolongs life in MDS, offering some hope and direction in the future management of this deadly disease.
Collapse
Affiliation(s)
- Jean-Pierre Issa
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA.
| |
Collapse
|
27
|
Gene-specific and global methylation patterns predict outcome in patients with acute myeloid leukemia. Leukemia 2010; 24:932-41. [DOI: 10.1038/leu.2010.41] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
28
|
Smith BD, Karp JE. What are the endpoints of therapy for acute leukemias? Old definitions and new challenges. ACTA ACUST UNITED AC 2010; 9 Suppl 3:S296-301. [PMID: 19778856 DOI: 10.3816/clm.2009.s.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Acute leukemias are complex diseases on multiple levels, and laboratory efforts over the past 3 decades have focused on better understanding of the molecular underpinnings and their stem cell biology. We now have a panoply of technologic advances that allow us to characterize individual leukemias by molecular profiles that relate directly to clinical behavior, to detect minimal residual disease, and to begin to develop "targeted" therapeutic strategies based on molecular considerations. There are a number of challenges surrounding this task: first, how to combine these agents with traditional chemotherapeutics and/or with each other to maximize leukemic cell kill and increase the cure rate; second, how to use these targeted agents in the minimal residual disease with potential curative intent; third, for patients unable to tolerate or unlikely to benefit from aggressive approaches, how to use one or more of these agents to reduce tumor bulk and either permit some restoration of normal marrow function or induce morphologic and functional differentiation of the leukemic clone to overcome the leukemia-associated bone marrow failure; and lastly, how to measure the effects of these agents on the molecular and cellular biologic levels in ways that correlate with and might even predict overall clinical outcome. These challenges are further complicated by the inherent heterogeneity in host biology; disease etiology and biology; and interactions among host, disease, and treatment that ultimately determine individual clinical outcomes. Toward this end, we will discuss selected issues surrounding new clinical trial designs and the development of clinically relevant molecular endpoints that might facilitate the development of new treatment approaches that will improve the outlook for adults with acute leukemias.
Collapse
Affiliation(s)
- B Douglas Smith
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21231-1000, USA
| | | |
Collapse
|
29
|
Santos FPS, Kantarjian H, Garcia-Manero G, Issa JP, Ravandi F. Decitabine in the treatment of myelodysplastic syndromes. Expert Rev Anticancer Ther 2010; 10:9-22. [PMID: 20014881 DOI: 10.1586/era.09.164] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Myelodysplastic syndromes (MDS) are a group of heterogeneous clonal hematopoietic stem cell disorders characterized by ineffective hematopoiesis, peripheral blood cytopenias and a propensity to transform into acute myeloid leukemia. There are few treatment options available for patients with MDS. Studies into the molecular biology of MDS have demonstrated abnormal patterns of DNA methylation that lead to silencing of tumor-suppressor genes. Hypomethylating agents are compounds that have the potential to reverse the aberrant DNA methylation and increase the expression of silenced genes, leading to cellular differentiation and/or apoptosis. Decitabine is a cytidine analogue that has activity as a hypomethylating agent and has been evaluated in the therapy of patients with high-risk MDS. Several studies have confirmed the clinical activity of low-dose decitabine in patients with high-risk MDS, leading to responses in approximately 50% of patients, with low treatment-related mortality. Responses have even been seen in patients with high-risk cytogenetic abnormalities, and some studies have demonstrated increased re-expression of genes that were previously silenced by hypermethylation, such as CDKN2B/p15INK4B. There are still some issues concerning the ideal dose and schedule of decitabine for treating patients with MDS. This article focuses on the most recent clinical studies of decitabine for therapy of MDS.
Collapse
Affiliation(s)
- Fabio P S Santos
- Department of Leukemia, University of Texas, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 0428, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
30
|
Shen L, Kantarjian H, Guo Y, Lin E, Shan J, Huang X, Berry D, Ahmed S, Zhu W, Pierce S, Kondo Y, Oki Y, Jelinek J, Saba H, Estey E, Issa JPJ. DNA methylation predicts survival and response to therapy in patients with myelodysplastic syndromes. J Clin Oncol 2009; 28:605-13. [PMID: 20038729 DOI: 10.1200/jco.2009.23.4781] [Citation(s) in RCA: 280] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PURPOSE The current classification systems of myelodysplastic syndromes (MDS), including the International Prognostic Scoring System (IPSS), do not fully reflect the molecular heterogeneity of the disease. Molecular characterization may predict clinical outcome and help stratify patients for targeted therapies. Epigenetic therapy using decitabine, a DNA hypomethylating agent, is clinically effective for the treatment of MDS. Therefore, we investigated the association between DNA methylation and clinical outcome in MDS. PATIENTS AND METHODS We screened 24 patients with MDS for promoter CpG island methylation of 24 genes and identified aberrant hypermethylation at 10 genes. We then performed quantitative methylation analyses by bisulfite pyrosequencing of the identified genes in 317 patient samples from three independent studies and assessed relations between methylation and clinical outcome. RESULTS In an initial training cohort of 89 patients with MDS, methylation frequencies of individual genes ranged from 7% to 70% and were highly concordant. Therefore, we defined a methylation z score based on all genes for each patient. We found that patients with higher levels of methylation, compared with patients with lower levels, had a shorter median overall survival (12.3 v 17.5 months, respectively; P = .04) and shorter median progression-free survival (6.4 v 14.9 months, respectively; P = .009). This methylation prognostic model was independent of age, sex, and IPSS group. Applied to two validation cohorts (228 patients), this model was confirmed as an independent prognostic predictor for survival. Although methylation at baseline did not correlate with clinical response to decitabine, we observed a significant correlation between reduced methylation over time and clinical responses. CONCLUSION DNA methylation predicts overall and progression-free survival in MDS.
Collapse
Affiliation(s)
- Lanlan Shen
- Department of Leukemia and Biostatistics, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Voso MT, Santini V, Finelli C, Musto P, Pogliani E, Angelucci E, Fioritoni G, Alimena G, Maurillo L, Cortelezzi A, Buccisano F, Gobbi M, Borin L, Di Tucci A, Zini G, Petti MC, Martinelli G, Fabiani E, Fazi P, Vignetti M, Piciocchi A, Liso V, Amadori S, Leone G. Valproic acid at therapeutic plasma levels may increase 5-azacytidine efficacy in higher risk myelodysplastic syndromes. Clin Cancer Res 2009; 15:5002-7. [PMID: 19638460 DOI: 10.1158/1078-0432.ccr-09-0494] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Epigenetic changes play a role and cooperate with genetic alterations in the pathogenesis of myelodysplastic syndromes (MDS). We conducted a phase II multicenter study on the combination of the DNA-methyltransferase inhibitor 5-azacytidine (5-AZA) and the histone deacetylase inhibitor valproic acid (VPA) in patients with higher risk MDS. EXPERIMENTAL DESIGN We enrolled 62 patients with MDS (refractory anemia with excess blasts, 39 patients; refractory anemia with excess blasts in transformation, 19 patients; and chronic myelomanocytic leukemia (CMML), 4 patients) and an International Prognostic Scoring System (IPSS) rating of Intermediate-2 (42 patients) or high (20 patients). VPA was given to reach a plasma concentration of >50 microg/mL, then 5-AZA was added s.c. at 75 mg/m(2) for 7 days in eight monthly cycles. RESULTS The median overall survival was 14.4 months. At a median follow-up of 12 months (range, 0.7-21.0), the disease progressed in 20 patients, with 21% cumulative incidence of progression. Of 26 patients who completed eight cycles, 30.7% obtained complete or partial remission, 15.4% had a major hematologic improvement, whereas 38.5% showed stable disease. Drug-related toxicity was mild. Favorable prognostic factors for survival were IPSS Intermediate-2 and plasma VPA of > or =50 microg/mL (log rank = 0.013 and 0.007, respectively). Analysis of polymorphisms important for the metabolism of the drugs used in the trial showed that carriers of the CYP2C19*2 variant of cytochrome P450 required higher VPA doses to achieve the target VPA plasma concentration of 50 microg/mL on day 1 of 5-AZA treatment (P = 0.0021). CONCLUSION Our data show that the 5-AZA/VPA combination is active and safe in patients with MDS with a poor prognosis. Achievement of VPA therapeutic levels may indeed increase 5-AZA efficacy.
Collapse
|
32
|
Abstract
Optimal management of patients with myelodysplastic syndromes (MDS) requires an insight into the biology of the disease and the mechanisms of action of the available therapies. This review focuses on low-risk MDS, for which chronic anaemia and eventual progression to acute myeloid leukaemia are the main concerns. We cover the updated World Health Organization classification, the latest prognostic scoring system, and describe novel findings in the pathogenesis of 5q- syndrome. We perform in depth analyses of two of the most widely used treatments, erythropoietin and lenalidomide, discussing mechanisms of action, reasons for treatment failure and influence on survival.
Collapse
Affiliation(s)
- M Jädersten
- Division of Hematology and Center of Experimental Hematology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | | |
Collapse
|
33
|
Candiloro ILM, Mikeska T, Hokland P, Dobrovic A. Rapid analysis of heterogeneously methylated DNA using digital methylation-sensitive high resolution melting: application to the CDKN2B (p15) gene. Epigenetics Chromatin 2008; 1:7. [PMID: 19014416 PMCID: PMC2590600 DOI: 10.1186/1756-8935-1-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2008] [Accepted: 11/03/2008] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Methylation-sensitive high resolution melting (MS-HRM) methodology is able to recognise heterogeneously methylated sequences by their characteristic melting profiles. To further analyse heterogeneously methylated sequences, we adopted a digital approach to MS-HRM (dMS-HRM) that involves the amplification of single templates after limiting dilution to quantify and to determine the degree of methylation. We used this approach to study methylation of the CDKN2B (p15) cell cycle progression inhibitor gene which is inactivated by DNA methylation in haematological malignancies of the myeloid lineage. Its promoter region usually shows heterogeneous methylation and is only rarely fully methylated. The methylation status of CDKN2B can be used as a biomarker of response to treatment. Therefore the accurate characterisation of its methylation is desirable. RESULTS MS-HRM was used to assess CDKN2B methylation in acute myeloid leukaemia (AML) samples. All the AML samples that were methylated at the CDKN2B promoter (40/93) showed varying degrees of heterogeneous methylation. Six representative samples were selected for further study. dMS-HRM was used to simultaneously count the methylated alleles and assess the degree of methylation. Direct sequencing of selected dMS-HRM products was used to determine the exact DNA methylation pattern and confirmed the degree of methylation estimated by dMS-HRM. CONCLUSION dMS-HRM is a powerful technique for the analysis of methylation in CDKN2B and other heterogeneously methylated genes. It eliminates both PCR and cloning bias towards either methylated or unmethylated DNA. Potentially complex information is simplified into a digital output, allowing counting of methylated and unmethylated alleles and providing an overall picture of methylation at the given locus. Downstream sequencing is minimised as dMS-HRM acts as a screen to select only methylated clones for further analysis.
Collapse
Affiliation(s)
- Ida L M Candiloro
- Molecular Pathology Research and Development Laboratory, Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria 8006, Australia.
| | | | | | | |
Collapse
|
34
|
Abstract
The goal of personalized medicine is to recommend drug treatment based on an individual's genetic makeup. Pharmacogenomic studies utilize two main approaches: candidate gene and whole-genome. Both approaches analyze genetic variants such as single nucleotide polymorphisms (SNPs) to identify associations with drug response. In addition to DNA sequence variations, non-genetic but heritable epigenetic systems have also been implicated in regulating gene expression that could influence drug response. The International HapMap Project lymphoblastoid cell lines (LCLs) have been used to study genetic determinants responsible for expression variation and drug response. Recent studies have demonstrated that common genetic variants, including both SNPs and copy number variants (CNVs) account for a substantial fraction of natural variation in gene expression. Given the critical role played by DNA methylation in gene regulation and the fact that DNA methylation is currently the most studied epigenetic system, we suggest that profiling the variation in DNA methylation in the HapMap samples will provide new insights into the regulation of gene expression as well as the mechanisms of individual drug response at a new level of complexity. Epigenomics will substantially add to our knowledge of how genetics explains gene expression and pharmacogenomics.
Collapse
Affiliation(s)
- Wei Zhang
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - R Stephanie Huang
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - M Eileen Dolan
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
35
|
Abstract
Myelodysplastic syndromes (MDSs) are clonal hematologic disorders that frequently represent an intermediate disease stage before progression to acute myeloid leukemia (AML). As such, study of MDS/AML can provide insight into the mechanisms of neoplastic evolution. In 184 patients with MDS and AML, DNA methylation microarray and high-density single nucleotide polymorphism array (SNP-A) karyotyping were used to assess the relative contributions of aberrant DNA methylation and chromosomal deletions to tumor-suppressor gene (TSG) silencing during disease progression. Aberrant methylation was seen in every sample, on average affecting 91 of 1505 CpG loci in early MDS and 179 of 1505 loci after blast transformation (refractory anemia with excess blasts [RAEB]/AML). In contrast, chromosome aberrations were seen in 79% of early MDS samples and 90% of RAEB/AML samples, and were not as widely distributed over the genome. Analysis of the most frequently aberrantly methylated genes identified FZD9 as a candidate TSG on chromosome 7. In patients with chromosome deletion at the FZD9 locus, aberrant methylation of the remaining allele was associated with the poorest clinical outcome. These results indicate that aberrant methylation can cooperate with chromosome deletions to silence TSG. However, the ubiquity, extent, and correlation with disease progression suggest that aberrant DNA methylation is the dominant mechanism for TSG silencing and clonal variation in MDS evolution to AML.
Collapse
|
36
|
Kantarjian H, O'Brien S, Cortes J, Wierda W, Faderl S, Garcia-Manero G, Issa JP, Estey E, Keating M, Freireich EJ. Therapeutic advances in leukemia and myelodysplastic syndrome over the past 40 years. Cancer 2008; 113:1933-52. [PMID: 18798533 PMCID: PMC4392892 DOI: 10.1002/cncr.23655] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Major therapeutic progress has been accomplished in leukemia and myelodysplastic syndrome (MDS) over the past 40 years, which may not be fully appreciated by the larger medical community. The objective of this review was to briefly highlight the treatment breakthroughs in leukemia and MDS. Therapeutic progress happened through better understanding of disease pathophysiologies and rational development of targeted agents, like imatinib mesylate in chronic myeloid leukemia (CML), and through astute, empirical discoveries in the clinic, like all-trans retinoic acid and arsenic trioxide in acute promyelocytic leukemia (APL) and chlorodeoxyadenosine in hairy cell leukemia (HCL). Today, the 5- to 10-year survival rates in patients with APL and HCL exceed 80%. In patients with CML, imatinib therapy has been associated with estimated 5- to 7-year survival rates from 85% to 90%. In patients with adult acute lymphocytic leukemia, modern intensive regimens have improved the 5-year survival rates from 20% up to 40%. In patients with chronic lymphocytic leukemia, chemoimmunotherapy recently produced high rates of quality responses and improved long-term outcome. In younger patients with acute myeloid leukemia (AML), the 5-year survival rates range from 40% to 50%, although elderly AML remains a therapeutic challenge. In patients with MDS, it was recently demonstrated that epigenetic therapy with hypomethylating agents improved survival. Much therapeutic progress has been witnessed in leukemia and MDS, and much more is expected to occur soon.
Collapse
MESH Headings
- Humans
- Leukemia/mortality
- Leukemia/therapy
- Leukemia, Hairy Cell/mortality
- Leukemia, Hairy Cell/therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/therapy
- Myelodysplastic Syndromes/mortality
- Myelodysplastic Syndromes/therapy
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy
Collapse
Affiliation(s)
- Hagop Kantarjian
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Flohr H, Breull W. Effect of etafenone on total and regional myocardial blood flow. Semin Hematol 1976; 50:16-37. [PMID: 23 DOI: 10.1053/j.seminhematol.2013.01.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The distribution of blood flow to the subendocardial, medium and subepicardial layers of the left ventricular free wall was studied in anaesthetized dogs under normoxic (A), hypoxic (B) conditions and under pharmacologically induced (etafenone) coronary vasodilation (C). Regional myocardial blood flow was determined by means of the particle distribution method. In normoxia a transmural gradient of flow was observed, with the subendocardial layers receiving a significantly higher flow rate compared with the subepicardial layers. In hypoxia induced vasodilation this transmural gradient of flow was persistent. In contrast a marked redistribution of regional flow was observed under pharmacologically induced vasodilation. The transmural gradient decreased. In contrast to some findings these experiments demonstrate that a considerable vasodilatory capacity exists in all layers of the myocardium and can be utilized by drugs. The differences observed for the intramural distribution pattern of flow under hypoxia and drug induced vasodilation support the hypothesis that this pattern reflects corresponding gradients of regional myocardial metabolism.
Collapse
|