1
|
Dang T, Guan X, Cui L, Ruan Y, Chen Z, Zou H, Lan Y, Liu C, Zhang Y. Epigenetics and immunotherapy in colorectal cancer: progress and promise. Clin Epigenetics 2024; 16:123. [PMID: 39252116 PMCID: PMC11385519 DOI: 10.1186/s13148-024-01740-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 09/02/2024] [Indexed: 09/11/2024] Open
Abstract
Colorectal cancer (CRC) is a common malignant tumor with the third and second highest incidence and mortality rates among various malignant tumors. Despite significant advancements in the present therapy for CRC, the majority of CRC cases feature proficient mismatch repair/microsatellite stability and have no response to immunotherapy. Therefore, the search for new treatment options holds immense importance in the diagnosis and treatment of CRC. In recent years, clinical research on immunotherapy combined with epigenetic therapy has gradually increased, which may bring hope for these patients. This review explores the role of epigenetic regulation in exerting antitumor effects through its action on immune cell function and highlights the potential of certain epigenetic genes that can be used as markers of immunotherapy to predict therapeutic efficacy. We also discuss the application of epigenetic drug sensitization immunotherapy to develop new treatment options combining epigenetic therapy and immunotherapy.
Collapse
Affiliation(s)
- Tianjiao Dang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150001, Heilongjiang, People's Republic of China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China
| | - Xin Guan
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150001, Heilongjiang, People's Republic of China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China
| | - Luying Cui
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150001, Heilongjiang, People's Republic of China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China
| | - Yuli Ruan
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150001, Heilongjiang, People's Republic of China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China
| | - Zhuo Chen
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Haoyi Zou
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150001, Heilongjiang, People's Republic of China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China
| | - Ya Lan
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Chao Liu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150001, Heilongjiang, People's Republic of China.
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China.
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China.
| | - Yanqiao Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150001, Heilongjiang, People's Republic of China.
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China.
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China.
| |
Collapse
|
2
|
Chiou JT, Chang LS. Synergistic cytotoxicity of decitabine and YM155 in leukemia cells through upregulation of SLC35F2 and suppression of MCL1 and survivin expression. Apoptosis 2024; 29:503-520. [PMID: 38066391 DOI: 10.1007/s10495-023-01918-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2023] [Indexed: 02/18/2024]
Abstract
The hypomethylation agent decitabine (DAC), in combination with other apoptosis inducers, is considered a potential modality for cancer treatment. We investigated the mechanism underlying the combined cytotoxicity of DAC and YM155 in acute myeloid leukemia (AML) cells because of increasing evidence that YM155 induces apoptosis in cancer cells. Co-administration of DAC and YM155 resulted in synergistic cytotoxicity in AML U937 cells, which was characterized by the induction of apoptosis, NOXA-dependent degradation of MCL1 and survivin, and depolarization of mitochondria. Restoration of MCL1 or survivin expression attenuated DAC/YM155-induced U937 cell death. DAC initiated AKT and p38 MAPK phosphorylation in a Ca2+/ROS-dependent manner, thereby promoting autophagy-mediated degradation of β-TrCP mRNA, leading to increased Sp1 expression. DAC-induced Sp1 expression associated with Ten-eleven-translocation (TET) dioxygenases and p300 was used to upregulate the expression of SLC35F2. Simultaneously, the activation of p38 MAPK induced by DAC, promoted CREB-mediated NOXA expression, resulting in survivin and MCL1 degradation. The synergistic cytotoxicity of DAC and YM155 in U937 cells was dependent on elevated SLC35F2 expression. Additionally, YM155 facilitated DAC-induced degradation of MCL1 and survivin. A similar mechanism explained DAC/YM155-mediated cytotoxicity in AML HL-60 cells. Our data demonstrated that the synergistic cytotoxicity of DAC and YM155 in AML cell lines U937 and HL-60 is dependent on AKT- and p38 MAPK-mediated upregulation of SLC35F2 and p38 MAPK-mediated degradation of survivin and MCL1. This indicates that a treatment regimen that amalgamates YM155 and DAC may be beneficial for AML.
Collapse
Affiliation(s)
- Jing-Ting Chiou
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
| | - Long-Sen Chang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan.
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| |
Collapse
|
3
|
Xu Y, Li P, Liu Y, Xin D, Lei W, Liang A, Han W, Qian W. Epi-immunotherapy for cancers: rationales of epi-drugs in combination with immunotherapy and advances in clinical trials. Cancer Commun (Lond) 2022; 42:493-516. [PMID: 35642676 PMCID: PMC9198339 DOI: 10.1002/cac2.12313] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 03/03/2022] [Accepted: 05/18/2022] [Indexed: 11/12/2022] Open
Abstract
Over the last two decades, several epi-drugs, immune checkpoint inhibitors (ICIs) and adoptive cell therapies have received clinical approval for use in certain types of cancer. However, monotherapy with epi-drugs or ICIs has shown limited efficacy in most cancer patients. Epigenetic agents have been shown to regulate the crosstalk between the tumor and host immunity to alleviate immune evasion, suggesting that epi-drugs can potentially synergize with immunotherapy. In this review, we discuss recent insights into the rationales of incorporating epigenetic therapy into immunotherapy, called epi-immunotherapy, and focus on an update of current clinical trials in both hematological and solid malignancies. Furthermore, we outline the future challenges and strategies in the field of cancer epi-immunotherapy.
Collapse
Affiliation(s)
- Yang Xu
- Department of Hematologythe Second Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouZhejiang310009P. R. China
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell BiologyLife Sciences InstituteZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Ping Li
- Department of HematologyTongji Hospital of Tongji UniversityShanghai200065P. R. China
| | - Yang Liu
- Department of Bio‐Therapeuticthe First Medical CentreChinese PLA General HospitalBeijing100853P. R. China
| | - Dijia Xin
- Department of Hematologythe Second Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouZhejiang310009P. R. China
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell BiologyLife Sciences InstituteZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Wen Lei
- Department of Hematologythe Second Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouZhejiang310009P. R. China
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell BiologyLife Sciences InstituteZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Aibin Liang
- Department of HematologyTongji Hospital of Tongji UniversityShanghai200065P. R. China
| | - Weidong Han
- Department of Bio‐Therapeuticthe First Medical CentreChinese PLA General HospitalBeijing100853P. R. China
| | - Wenbin Qian
- Department of Hematologythe Second Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouZhejiang310009P. R. China
| |
Collapse
|
4
|
Fang DD, Tang Q, Kong Y, Rong T, Wang Q, Li N, Fang X, Gu J, Xiong D, Yin Y, Deng J, Yang D, Zhai Y. MDM2 inhibitor APG-115 exerts potent antitumor activity and synergizes with standard-of-care agents in preclinical acute myeloid leukemia models. Cell Death Discov 2021; 7:90. [PMID: 33941774 PMCID: PMC8093284 DOI: 10.1038/s41420-021-00465-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 02/28/2021] [Accepted: 03/21/2021] [Indexed: 12/31/2022] Open
Abstract
Acute myeloid leukemia (AML) is a clinically and genetically heterogeneous clonal disease associated with unmet medical needs. Paralleling the pathology of other cancers, AML tumorigenesis and propagation can be ascribed to dysregulated cellular processes, including apoptosis. This function and others are regulated by tumor suppressor P53, which plays a pivotal role in leukemogenesis. Opposing P53-mediated activities is the mouse double minute 2 homolog (MDM2), which promotes P53 degradation. Because the TP53 mutation rate is low, and MDM2 frequently overexpressed, in patients with leukemia, targeting the MDM2-P53 axis to restore P53 function has emerged as an attractive AML treatment strategy. APG-115 is a potent MDM2 inhibitor under clinical development for patients with solid tumors. In cellular cultures and animal models of AML, we demonstrate that APG-115 exerted substantial antileukemic activity, as either a single agent or when combined with standard-of-care (SOC) hypomethylating agents azacitidine (AZA) and decitabine (DAC), or the DNA-damaging agent cytarabine (Ara-C). By activating the P53/P21 pathway, APG-115 exhibited potent antiproliferative and apoptogenic activities, and induced cell cycle arrest, in TP53 wild-type AML lines. In vivo, APG-115 significantly reduced tumor burden and prolonged survival. Combinations of APG-115 with SOC treatments elicited synergistic antileukemic activity. To explain these effects, we propose that APG-115 and SOC agents augment AML cell killing by complementarily activating the P53/P21 pathway and upregulating DNA damage. These findings and the emerging mechanism of action afford a sound scientific rationale to evaluate APG-115 (with or without SOC therapies) in patients with AML.
Collapse
Affiliation(s)
- Douglas D Fang
- Ascentage Pharma (Suzhou) Co., Ltd., 218 Xinghu Street, Suzhou, Jiangsu Province, China
| | - Qiuqiong Tang
- Ascentage Pharma (Suzhou) Co., Ltd., 218 Xinghu Street, Suzhou, Jiangsu Province, China
| | - Yanhui Kong
- Ascentage Pharma (Suzhou) Co., Ltd., 218 Xinghu Street, Suzhou, Jiangsu Province, China
| | - Tao Rong
- Ascentage Pharma (Suzhou) Co., Ltd., 218 Xinghu Street, Suzhou, Jiangsu Province, China
| | - Qixin Wang
- Ascentage Pharma (Suzhou) Co., Ltd., 218 Xinghu Street, Suzhou, Jiangsu Province, China
| | - Na Li
- Ascentage Pharma (Suzhou) Co., Ltd., 218 Xinghu Street, Suzhou, Jiangsu Province, China
| | - Xu Fang
- Ascentage Pharma (Suzhou) Co., Ltd., 218 Xinghu Street, Suzhou, Jiangsu Province, China
| | - Jiaxing Gu
- Ascentage Pharma (Suzhou) Co., Ltd., 218 Xinghu Street, Suzhou, Jiangsu Province, China
| | - Dengkun Xiong
- Ascentage Pharma (Suzhou) Co., Ltd., 218 Xinghu Street, Suzhou, Jiangsu Province, China
| | - Yan Yin
- Ascentage Pharma (Suzhou) Co., Ltd., 218 Xinghu Street, Suzhou, Jiangsu Province, China
| | - Jing Deng
- Ascentage Pharma (Suzhou) Co., Ltd., 218 Xinghu Street, Suzhou, Jiangsu Province, China
| | - Dajun Yang
- Ascentage Pharma (Suzhou) Co., Ltd., 218 Xinghu Street, Suzhou, Jiangsu Province, China.
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| | - Yifan Zhai
- Ascentage Pharma (Suzhou) Co., Ltd., 218 Xinghu Street, Suzhou, Jiangsu Province, China.
| |
Collapse
|
5
|
Kim MJ, Chen G, Sica GL, Deng X. Epigenetic modulation of FBW7/Mcl-1 pathway for lung cancer therapy. Cancer Biol Ther 2021; 22:55-65. [PMID: 33336620 PMCID: PMC7833779 DOI: 10.1080/15384047.2020.1856756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/24/2020] [Accepted: 11/23/2020] [Indexed: 01/09/2023] Open
Abstract
Methylation induces epigenetic silencing of tumor suppressor genes in human lung cancer. Inhibition of DNA methyltransferases by decitabine (DAC) can demethylate and activate epigenetically silenced tumor suppressor genes. Epigenetic therapy using DAC should be an attractive strategy for lung cancer therapy. FBW7 is a tumor suppressor that functions as an Mcl-1 E3 ligase to degrade Mcl-1 by ubiquitination. Here we discovered that treatment of various human lung cancer cells with DAC resulted in activation of FBW7 expression, decreased levels of Mcl-1 protein, and growth inhibition. DAC-activated FBW7 expression promoted Mcl-1 ubiquitination and degradation leading to a significant reduction in the half-life of Mcl-1 protein. Mechanistically, treatment of lung cancer cells or lung cancer xenografts with DAC induced the conversion of the FBW7 gene from a methylated form to an unmethylated form, which was associated with the increased expression of FBW7 and decreased expression of Mcl-1 in vitro and in vivo. DAC suppressed lung cancer growth in a dose-dependent manner in vivo. Combined treatment with DAC and a Bcl2 inhibitor, venetoclax, exhibited strong synergistic potency against lung cancer without normal tissue toxicity. These findings uncover a novel mechanism by which DAC suppresses tumor growth by targeting the FBW7/Mcl-1 signaling pathway. Combination of DAC with Bcl2 inhibitor venetoclax provides more effective epigenetic therapy for lung cancer.
Collapse
Affiliation(s)
- Mi Jeong Kim
- Department of Radiation Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, Georgia, USA
- Department of Food and Nutrition, Changwon National University, Gyeongsangnam-do, Korea
| | - Guo Chen
- Department of Radiation Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, Georgia, USA
| | - Gabriel L. Sica
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, Georgia, USA
| | - Xingming Deng
- Department of Radiation Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, Georgia, USA
| |
Collapse
|
6
|
Liu Y, Mondello P, Erazo T, Tannan NB, Asgari Z, de Stanchina E, Nanjangud G, Seshan VE, Wang S, Wendel HG, Younes A. NOXA genetic amplification or pharmacologic induction primes lymphoma cells to BCL2 inhibitor-induced cell death. Proc Natl Acad Sci U S A 2018; 115:12034-12039. [PMID: 30404918 PMCID: PMC6255185 DOI: 10.1073/pnas.1806928115] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Although diffuse large B cell lymphoma (DLBCL) cells widely express the BCL2 protein, they rarely respond to treatment with BCL2-selective inhibitors. Here we show that DLBCL cells harboring PMAIP1/NOXA gene amplification were highly sensitive to BCL2 small-molecule inhibitors. In these cells, BCL2 inhibition induced cell death by activating caspase 9, which was further amplified by caspase-dependent cleavage and depletion of MCL1. In DLBCL cells lacking NOXA amplification, BCL2 inhibition was associated with an increase in MCL1 protein abundance in a BIM-dependent manner, causing a decreased antilymphoma efficacy. In these cells, dual inhibition of MCL1 and BCL2 was required for enhanced killing. Pharmacologic induction of NOXA, using the histone deacetylase inhibitor panobinostat, decreased MCL1 protein abundance and increased lymphoma cell vulnerability to BCL2 inhibitors in vitro and in vivo. Our data provide a mechanistic rationale for combination strategies to disrupt lymphoma cell codependency on BCL2 and MCL1 proteins in DLBCL.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Apoptosis Regulatory Proteins/metabolism
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Female
- Gene Amplification/drug effects
- Histone Deacetylase Inhibitors/pharmacology
- Histone Deacetylase Inhibitors/therapeutic use
- Humans
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- Mice
- Mice, Nude
- Myeloid Cell Leukemia Sequence 1 Protein/metabolism
- Panobinostat/pharmacology
- Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Yuxuan Liu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Patrizia Mondello
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Tatiana Erazo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Neeta Bala Tannan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Zahra Asgari
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Elisa de Stanchina
- Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Gouri Nanjangud
- Molecular Cytogenetics Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Venkatraman E Seshan
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Shenqiu Wang
- Cancer Biology and Genetics Program, Sloan Kettering Institute for Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Hans-Guido Wendel
- Cancer Biology and Genetics Program, Sloan Kettering Institute for Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Anas Younes
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065;
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| |
Collapse
|
7
|
Jiang C, Gong F. MiR-148a promotes myocardial differentiation of human bone mesenchymal stromal cells via DNA methyltransferase 1 (DNMT1). Cell Biol Int 2018; 42:913-922. [PMID: 28656724 DOI: 10.1002/cbin.10813] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 06/24/2017] [Indexed: 11/08/2022]
Abstract
MicroRNAs have potential to modulate the differentiation of stem cells. In previous study, we found that miR-148a was up-regulated in myocardial differentiation of human bone mesenchymal stromal cells (hBMSCs) induced by 5'-azacytidine. However, the role of miR-148a in regulating this process still remains unclear. In this study, we investigated the function and molecular mechanism of miR-148a in myocardial differentiation of hBMSCs. We found that miR-148a was significantly increased while DNA methyltransferase 1 (DNMT1) was significantly decreased in myocardial differentiation of hBMSCs. Then, the dual luciferase reporter assays method indicated that DNMT1 was the direct target of miR-148a. In addition, we showed that up-regulation of miR-148a could enhance myocardial differentiation of hBMSCs, while down-regulation of miR-148a could inhibit myocardial differentiation process. Moreover, knockdown of DNMT1 could block the role of miR-148a in promoting myocardial differentiation of hBMSCs. Finally, MiR-148a acted on methylation level of GATA-4 and knockdown of DNMT1 could block this function. Therefore, our results indicate that miR-148a plays a vital role in regulating myocardial differentiation of hBMSCs by targeting DNMT1.
Collapse
Affiliation(s)
- Changke Jiang
- Department of Pediatrics, Yongchuan Hospital of Chongqing Medical University, 439 Xuanhua Road, Yongchuan, Chongqing, 402160, China
| | - Fang Gong
- Department of Pediatrics, Yongchuan Hospital of Chongqing Medical University, 439 Xuanhua Road, Yongchuan, Chongqing, 402160, China
| |
Collapse
|
8
|
Gene and MicroRNA Perturbations of Cellular Response to Pemetrexed Implicate Biological Networks and Enable Imputation of Response in Lung Adenocarcinoma. Sci Rep 2018; 8:733. [PMID: 29335598 PMCID: PMC5768793 DOI: 10.1038/s41598-017-19004-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 12/20/2017] [Indexed: 12/18/2022] Open
Abstract
Pemetrexed is indicated for non-small cell lung carcinoma and mesothelioma, but often has limited efficacy due to drug resistance. To probe the molecular mechanisms underlying chemotherapeutic response, we performed mRNA and microRNA (miRNA) expression profiling of pemetrexed treated and untreated lymphoblastoid cell lines (LCLs) and applied a hierarchical Bayesian method. We identified genetic variation associated with gene expression in human lung tissue for the most significant differentially expressed genes (Benjamini-Hochberg [BH] adjusted p < 0.05) using the Genotype-Tissue Expression data and found evidence for their clinical relevance using integrated molecular profiling and lung adenocarcinoma survival data from The Cancer Genome Atlas project. We identified 39 miRNAs with significant differential expression (BH adjusted p < 0.05) in LCLs. We developed a gene expression based imputation model of drug sensitivity, quantified its prediction performance, and found a significant correlation of the imputed phenotype generated from expression data with survival time in lung adenocarcinoma patients. Differentially expressed genes (MTHFD2 and SUFU) that are putative targets of differentially expressed miRNAs also showed differential perturbation in A549 fusion lung tumor cells with further replication in A549 cells. Our study suggests pemetrexed may be used in combination with agents that target miRNAs to increase its cytotoxicity.
Collapse
|
9
|
Young CS, Clarke KM, Kettyle LM, Thompson A, Mills KI. Decitabine-Vorinostat combination treatment in acute myeloid leukemia activates pathways with potential for novel triple therapy. Oncotarget 2017; 8:51429-51446. [PMID: 28881658 PMCID: PMC5584259 DOI: 10.18632/oncotarget.18009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 05/07/2017] [Indexed: 01/21/2023] Open
Abstract
Despite advancements in cancer therapeutics, acute myeloid leukemia patients over 60 years old have a 5-year survival rate of less than 8%. In an attempt to improve this, epigenetic modifying agents have been combined as therapies in clinical studies. In particular combinations with Decitabine and Vorinostat have had varying degrees of efficacy. This study therefore aimed to understand the underlying molecular mechanisms of these agents to identify potential rational epi-sensitized combinations. Combined Decitabine-Vorinostat treatment synergistically decreased cell proliferation, induced apoptosis, enhanced acetylation of histones and further decreased DNMT1 protein with HL-60 cells showing a greater sensitivity to the combined treatment than OCI-AML3. Combination therapy led to reprogramming of unique target genes including AXL, a receptor tyrosine kinase associated with cell survival and a poor prognosis in AML, which was significantly upregulated following treatment. Therefore targeting AXL following epi-sensitization with Decitabine and Vorinostat may be a suitable triple combination. To test this, cells were treated with a novel triple combination therapy including BGB324, an AXL specific inhibitor. Triple combination increased the sensitivity of OCI-AML3 cells to Decitabine and Vorinostat as shown through viability assays and significantly extended the survival of mice transplanted with pretreated OCI-AML3 cells, while bioluminescence imaging showed the decrease in disease burden following triple combination treatment. Further investigation is required to optimize this triple combination, however, these results suggest that AXL is a potential marker of response to Decitabine-Vorinostat combination treatment and offers a new avenue of epigenetic combination therapies for acute myeloid leukemia.
Collapse
Affiliation(s)
- Christine S. Young
- Blood Cancer Research Group, Centre for Cancer Research and Cell Biology, Queen's University, Belfast, United Kingdom
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | - Kathryn M. Clarke
- Blood Cancer Research Group, Centre for Cancer Research and Cell Biology, Queen's University, Belfast, United Kingdom
- Department of Haematology, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Laura M. Kettyle
- Blood Cancer Research Group, Centre for Cancer Research and Cell Biology, Queen's University, Belfast, United Kingdom
- Haematopoietic Stem Cell Biology Laboratory, MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, United Kingdom
| | - Alexander Thompson
- Blood Cancer Research Group, Centre for Cancer Research and Cell Biology, Queen's University, Belfast, United Kingdom
- Division of Cancer and Stem Cells, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Ken I. Mills
- Blood Cancer Research Group, Centre for Cancer Research and Cell Biology, Queen's University, Belfast, United Kingdom
| |
Collapse
|
10
|
Tortorella SM, Hung A, Karagiannis TC. The implication of cancer progenitor cells and the role of epigenetics in the development of novel therapeutic strategies for chronic myeloid leukemia. Antioxid Redox Signal 2015; 22:1425-62. [PMID: 25366930 DOI: 10.1089/ars.2014.6096] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
SIGNIFICANCE Chronic myeloid leukemia (CML) involves the malignant transformation of hematopoietic stem cells, defined largely by the Philadelphia chromosome and expression of the breakpoint cluster region-Abelson (BCR-ABL) oncoprotein. Pharmacological tyrosine kinase inhibitors (TKIs), including imatinib mesylate, have overcome limitations in conventional treatment for the improved clinical management of CML. RECENT ADVANCES Accumulated evidence has led to the identification of a subpopulation of quiescent leukemia progenitor cells with stem-like self renewal properties that may initiate leukemogenesis, which are also shown to be present in residual disease due to their insensitivity to tyrosine kinase inhibition. CRITICAL ISSUES The characterization of quiescent leukemia progenitor cells as a unique cell population in CML pathogenesis has become critical with the complete elucidation of mechanisms involved in their survival independent of BCR-ABL that is important in the development of novel anticancer strategies. Understanding of these functional pathways in CML progenitor cells will allow for their selective therapeutic targeting. In addition, disease pathogenesis and drug responsiveness is also thought to be modulated by epigenetic regulatory mechanisms such as DNA methylation, histone acetylation, and microRNA expression, with a capacity to control CML-associated gene transcription. FUTURE DIRECTIONS A number of compounds in combination with TKIs are under preclinical and clinical investigation to assess their synergistic potential in targeting leukemic progenitor cells and/or the epigenome in CML. Despite the collective promise, further research is required in order to refine understanding, and, ultimately, advance antileukemic therapeutic strategies.
Collapse
Affiliation(s)
- Stephanie M Tortorella
- 1 Epigenomic Medicine, Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct , Melbourne, Australia
| | | | | |
Collapse
|
11
|
Maes K, De Smedt E, Lemaire M, De Raeve H, Menu E, Van Valckenborgh E, McClue S, Vanderkerken K, De Bruyne E. The role of DNA damage and repair in decitabine-mediated apoptosis in multiple myeloma. Oncotarget 2015; 5:3115-29. [PMID: 24833108 PMCID: PMC4102796 DOI: 10.18632/oncotarget.1821] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
DNA methyltransferase inhibitors (DNMTi) and histone deacetylase inhibitors (HDACi) are under investigation for the treatment of cancer, including the plasma cell malignancy multiple myeloma (MM). Evidence exists that DNA damage and repair contribute to the cytotoxicity mediated by the DNMTi decitabine. Here, we investigated the DNA damage response (DDR) induced by decitabine in MM using 4 human MM cell lines and the murine 5T33MM model. In addition, we explored how the HDACi JNJ-26481585 affects this DDR. Decitabine induced DNA damage (gamma-H2AX foci formation), followed by a G0/G1- or G2/M-phase arrest and caspase-mediated apoptosis. JNJ-26481585 enhanced the anti-MM effect of decitabine both in vitro and in vivo. As JNJ-26481585 did not enhance decitabine-mediated gamma-H2AX foci formation, we investigated the DNA repair response towards decitabine and/or JNJ-26481585. Decitabine augmented RAD51 foci formation (marker for homologous recombination (HR)) and/or 53BP1 foci formation (marker for non-homologous end joining (NHEJ)). Interestingly, JNJ-26481585 negatively affected basal or decitabine-induced RAD51 foci formation. Finally, B02 (RAD51 inhibitor) enhanced decitabine-mediated apoptosis. Together, we report that decitabine-induced DNA damage stimulates HR and/or NHEJ. JNJ-26481585 negatively affects RAD51 foci formation, thereby providing an additional explanation for the combinatory effect between decitabine and JNJ-26481585.
Collapse
Affiliation(s)
- Ken Maes
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Zhan YS, Tan SW, Mao W, Jiang J, Liu HL, Wu B. Chemotherapy mediates intestinal injury via p53/p53 upregulated modulator of apoptosis (PUMA) signaling pathway. J Dig Dis 2014; 15:425-34. [PMID: 24814616 DOI: 10.1111/1751-2980.12157] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the potential mechanism and signaling pathway involved in chemotherapy-induced intestinal mucosal injury (CIMI), which is a common physiopathological problem in patients with cancer. METHODS For the in vivo experiment, mice received intraperitoneal injection of 5-fluorouracil (5-FU) at a dose of 75 mg/kg/day for 1, 3 or 5 days. Villus height and crypt depth of the small intestine, cell apoptosis and proliferation were then examined to determine the extent of CIMI. The expressions of Akt, p53, PUMA and p21 were evaluated both in vivo in mice models and in vitro in the IEC-6 and HCT116 cell lines. RESULTS After 5-FU therapy both the intestinal villus height (275.93 μm vs 164.52 μm, P < 0.001) and crypt depth (64.13 μm vs 42.48 μm, P < 0.001) were decreased. The apoptotic index was greatly increased from 0.32% to 15.84% (P < 0.001) and proliferation was suppressed (63.58% vs 39.15%, P < 0.001). Additionally, p53 expression was significantly increased in the intestinal crypt along with the expressions of PUMA and p21. Western blot showed that the administration of 5-FU induced p53/PUMA-mediated apoptosis and upregulated p21 expression to suppress cell proliferation. CONCLUSION Chemotherapy might mediate intestinal injury via p53/PUMA-mediated apoptotic signaling and the suppression of proliferation in response to p21.
Collapse
Affiliation(s)
- Ya Shi Zhan
- Department of Gastroenterology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | | | | | | | | | | |
Collapse
|
13
|
Combined treatment with low concentrations of decitabine and SAHA causes cell death in leukemic cell lines but not in normal peripheral blood lymphocytes. BIOMED RESEARCH INTERNATIONAL 2013; 2013:659254. [PMID: 24000324 PMCID: PMC3755446 DOI: 10.1155/2013/659254] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 06/28/2013] [Accepted: 07/13/2013] [Indexed: 12/15/2022]
Abstract
Epigenetic therapy reverting aberrant acetylation or methylation offers the possibility to target preferentially tumor cells and to preserve normal cells. Combination epigenetic therapy may further improve the effect of individual drugs. We investigated combined action of demethylating agent decitabine and histone deacetylase inhibitor SAHA (Vorinostat) on different leukemic cell lines in comparison with peripheral blood lymphocytes. Large decrease of viability, as well as huge p21WAF1 induction, reactive oxygen species formation, and apoptotic features due to combined decitabine and SAHA action were detected in leukemic cell lines irrespective of their p53 status, while essentially no effect was observed in response to the combined drug action in normal peripheral blood lymphocytes of healthy donors. p53-dependent apoptotic pathway was demonstrated to participate in the wtp53 CML-T1 leukemic cell line response, while significant influence of reactive oxygen species on viability decrease has been detected in p53-null HL-60 cell line.
Collapse
|
14
|
Abstract
In recent years, histone deacetylase inhibitors (HDACis), a novel class of agents that targets mechanistic abnormalities in cancers, have shown promising anti-cancer activity in both hematological and solid cancers. Among them, vorinostat was approved by FDA to treat cutaneous T-cell lymphoma and is being evaluated in other cancer types. Although initially designed to target histone deacetylase, vorinostat were found to have additional effects on other epigenetic machineries, for example acetylation of non-HDAC, methylation and microRNA (miRNA) expression. In this review, we examined all known mechanisms of action for vorinostat. We also summarized the current findings on the `crosstalk' between different epigenetic machineries. These findings suggest that improved understanding of epigenetic regulatory role of vorinostat and/or other HDACis will provide novel insights in improving utilization of this class of novel agents.
Collapse
Affiliation(s)
- Jean Lee
- Department of Medicine, University of Chicago, USA
| | | |
Collapse
|
15
|
Patties I, Kortmann RD, Glasow A. Inhibitory effects of epigenetic modulators and differentiation inducers on human medulloblastoma cell lines. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2013; 32:27. [PMID: 23672687 PMCID: PMC3666942 DOI: 10.1186/1756-9966-32-27] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 05/07/2013] [Indexed: 12/20/2022]
Abstract
Background Medulloblastoma (MB) is the most common malignant brain tumor in childhood with a 5-year survival of approximately 60%. We have recently shown that treatment of human MB cells with 5-aza-2’-deoxycytidine (5-aza-dC) reduces the clonogenic survival significantly. Here, we tested combinatorial effects of 5-aza-dC with other epigenetic (valproic acid, SAHA) and differentiation-inducing drugs (resveratrol, abacavir, retinoic acid) on human MB cells in vitro to intensify the antitumor therapy further. Methods Three human MB cell lines were treated with 5-aza-dC alone or in combination for three or six days. Metabolic activity was measured by WST-1 assay. To determine long-term reproductive survival, clonogenic assays were performed. Induction of DNA double-strand break (DSB) repair was measured by γH2AX assay. Results The applied single drugs, except for ATRA, reduced the metabolic activity dose-dependently in all MB cell lines. Longer treatment times enhanced the reduction of metabolic activity by 5-aza-dC. Combinatorial treatments showed differential, cell line-dependent responses indicating an important impact of the genetic background. 5-Aza-dC together with resveratrol was found to exert the most significant inhibitory effects on metabolic activity in all cell lines. 5-aza-dC alone reduced the clonogenicity of MB cells significantly and induced DSB with no further changes after adjuvant administration of resveratrol. Conclusion The observed significant decrease in metabolic activity by combinatorial treatment of MB cells with 5-aza-dC and resveratrol does not translate into long-term reproductive survival deficiency in vitro. Further studies in animal models are needed to clarify the resveratrol-mediated anticancer mechanisms in vivo.
Collapse
Affiliation(s)
- Ina Patties
- Department of Radiation Therapy, University of Leipzig, Stephanstraße 9a, Leipzig, 04103, Germany.
| | | | | |
Collapse
|
16
|
Abstract
p63 is a transcriptional factor implicated in cancer and development. The presence in TP63 gene of alternative promoters allows expression of one isoform containing the N-terminal transactivation domain (TA isoform) and one N-terminal truncated isoform (ΔN isoform). Complete ablation of all p63 isoforms produced mice with fatal developmental abnormalities, including lack of epidermal barrier, limbs and other epidermal appendages. Specific TAp63-null mice, although they developed normally, failed to undergo in DNA damage-induced apoptosis during primordial follicle meiotic arrest, suggesting a p63 involvement in maternal reproduction. Recent findings have elucidated the role in DNA damage response of a novel Hominidae p63 isoform, GTAp63, specifically expressed in human spermatic precursors. Thus, these findings suggest a unique strategy of p63 gene, to evolve in order to preserve the species as a guardian of reproduction. Elucidation of the biological basis of p63 function in reproduction may provide novel approaches to the control of human fertility.
Collapse
Affiliation(s)
- Ivano Amelio
- Medical Research Council; Toxicology Unit; Leicester University; Leicester, UK
- Department for Molecular Biomedical Research; VIB; Ghent University; Ghent, Belgium
- Department of Biomedical Molecular Biology; Ghent University; Ghent, Belgium
| | - Francesca Grespi
- Medical Research Council; Toxicology Unit; Leicester University; Leicester, UK
- Department for Molecular Biomedical Research; VIB; Ghent University; Ghent, Belgium
- Department of Biomedical Molecular Biology; Ghent University; Ghent, Belgium
| | | | - Gerry Melino
- Medical Research Council; Toxicology Unit; Leicester University; Leicester, UK
- Department for Molecular Biomedical Research; VIB; Ghent University; Ghent, Belgium
- Department of Biomedical Molecular Biology; Ghent University; Ghent, Belgium
- Biochemistry IDI-IRCCS Laboratory and Department of Experimental Medicine and Surgery; University of Rome “Tor Vergata;” Rome, Italy
| |
Collapse
|
17
|
Synergistic silencing by promoter methylation and reduced AP-2α transactivation of the proapoptotic HRK gene confers apoptosis resistance and enhanced tumor growth. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 182:84-95. [PMID: 23159945 DOI: 10.1016/j.ajpath.2012.09.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 09/06/2012] [Accepted: 09/18/2012] [Indexed: 02/05/2023]
Abstract
The Harakiri (HRK) gene encodes an important proapoptotic mitochondrial protein of the Bcl-2 family. HRK is expressed in normal tissues but is decreased in many cancers such as melanoma, the mechanisms of which have not been fully elucidated. Here, we demonstrate that HRK is silenced by hypermethylation of a major proximal CpG island in the HRK promoter. Furthermore, we show that HRK is a novel target gene regulated by the transcription factor AP-2α, which interacts with an AP-2α binding site in the HRK promoter. Hypermethylation of the major proximal CpG island (which contains the AP-2α binding site within the most densely methylated -218- to -194-bp region) inhibited AP-2α binding and transcriptional activity. Artificial overexpression of AP-2α in melanoma cells up-regulated HRK transcription, which was further restored by treatment with DNA methyltransferase inhibitor 5-azacytidine. Artificial overexpression of HRK by recombinant adenovirus induced caspase-dependent apoptosis, inhibited melanoma cell growth in vitro, and markedly reduced in vivo melanoma growth in a nude mouse xenograft model. RNA interference by siHRK or siAP-2α reversed the above effects. We conclude that the synergistic effects of HRK promoter hypermethylation and loss of AP-2α transactivation lead to HRK gene silencing and confer resistance to apoptosis and enhanced tumor growth. These novel molecular lesions may provide the basis for new therapeutic approaches to treating AP-2α- and HRK-deficient cancers.
Collapse
|
18
|
Scholz B, Marschalek R. Epigenetics and blood disorders. Br J Haematol 2012; 158:307-22. [DOI: 10.1111/j.1365-2141.2012.09193.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 05/09/2012] [Indexed: 12/25/2022]
Affiliation(s)
- Bastian Scholz
- Institute of Pharmaceutical Biology/DCAL; Biocentre; Goethe-University; Frankfurt/Main; Germany
| | - Rolf Marschalek
- Institute of Pharmaceutical Biology/DCAL; Biocentre; Goethe-University; Frankfurt/Main; Germany
| |
Collapse
|
19
|
Cervera E, Candelaria M, López-Navarro O, Labardini J, Gonzalez-Fierro A, Taja-Chayeb L, Cortes J, Gordillo-Bastidas D, Dueñas-González A. Epigenetic therapy with hydralazine and magnesium valproate reverses imatinib resistance in patients with chronic myeloid leukemia. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2012; 12:207-12. [PMID: 22420986 DOI: 10.1016/j.clml.2012.01.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 01/10/2012] [Accepted: 01/20/2012] [Indexed: 01/07/2023]
Abstract
UNLABELLED The epigenetic drugs hydralazine and valproate were administered in a compassionate manner to 8 patients with chronic myeloid leukemia (CML) refractory to imatinib. Two patients had a complete hematologic response (25%),1 major cytogenetic response, 1 complete cytogenetic response (25% any cytogenetic response), and 3 (37.5%)stable disease. No grade 3 or 4 toxicity was observed. These results show the ability of epigenetic therapy to revert imatinib resistance. BACKGROUND Epigenetic alterations participate in the development of acquired resistance to imatinib, hence, the DNA methylation, and histone deacetylase inhibitors hydralazine and valproate, respectively, has the potential to overcome it. PATIENT AND METHODS A series of 8 patients with chronic myeloid leukemia (CML) refractory to imatinib mesylate with no access to second-generation tyrosine kinase inhibitors were treated with hydralazine and valproate in a compassionate manner. Clinical efficacy and safety of these drugs added to imatinib mesylate were evaluated. RESULTS Two patients were in the blast phase, 5 were in the accelerated phase, and 1 was in the chronic phase. All the patients continued with the same dose of imatinib that they had been receiving at the time of development of resistance, with a median dose of 600 mg daily (range, 400-800 mg). The median time from diagnosis of CML to the start of hydralazine and valproate was 53.6 months (range, 19-84 months). Two (25%) patients had a complete hematologic response, one (12.5%) had an major cytogenetic response, and one (12.5%) had a complete cytogenetic response. Three (37.5%) patients had stable disease, and only one (12.5%) patient failed to respond. At a median follow-up time of 18 months (range, 3-18 months), the median survival had not been reached, and the projected overall survival was 63%. All the patients had mild neurologic toxicity, including distal tremor and somnolence. No grade 3 or 4 toxicity was observed. CONCLUSIONS Our results suggest that the epigenetic drugs hydralazine and valproate revert the resistance to imatinib in patients with CML.
Collapse
MESH Headings
- Adult
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/adverse effects
- Antineoplastic Agents/therapeutic use
- Benzamides
- Drug Resistance, Neoplasm/genetics
- Epigenesis, Genetic/drug effects
- Female
- Follow-Up Studies
- Humans
- Hydralazine/administration & dosage
- Hydralazine/adverse effects
- Hydralazine/therapeutic use
- Imatinib Mesylate
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality
- Male
- Middle Aged
- Piperazines/administration & dosage
- Piperazines/adverse effects
- Piperazines/pharmacology
- Piperazines/therapeutic use
- Pyrimidines/administration & dosage
- Pyrimidines/adverse effects
- Pyrimidines/pharmacology
- Pyrimidines/therapeutic use
- Treatment Outcome
- Valproic Acid/administration & dosage
- Valproic Acid/adverse effects
- Valproic Acid/therapeutic use
Collapse
Affiliation(s)
- Eduardo Cervera
- Hematology Department, Instituto Nacional de Cancerología, México City, Mexico
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Generation of reactive oxygen species during apoptosis induced by DNA-damaging agents and/or histone deacetylase inhibitors. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2011; 2011:253529. [PMID: 21949898 PMCID: PMC3178180 DOI: 10.1155/2011/253529] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 07/15/2011] [Accepted: 07/26/2011] [Indexed: 01/08/2023]
Abstract
Reactive oxygen species play an important role in the process of apoptosis in many cell types. In this paper, we analyzed the role of ROS in DNA-damaging agents (actinomycin D or decitabine), which induced apoptosis of leukemia cell line CML-T1 and normal peripheral blood lymphocytes (PBL). The possibility of synergism with histone deacetylase inhibitors butyrate or SAHA is also reported. We found that in cancer cell line, ROS production significantly contributed to apoptosis triggering, while in normal lymphocytes treated by cytostatic or cytotoxic drugs, necrosis as well as apoptosis occurred and large heterogeneity of ROS production was measured. Combined treatment with histone deacetylase inhibitor did not potentiate actinomycin D action, whereas combination of decitabine and SAHA brought synergistic ROS generation and apoptotic features in CML cell line. Appropriate decrease of cell viability indicated promising therapeutic potential of this combination in CML, but side effects on normal PBL should be taken into attention.
Collapse
|