Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 16(2): 397-407, 2017 02.
Article in English | MEDLINE | ID: mdl-27980103

ABSTRACT

Epigenetic drugs, such as DNA methylation inhibitors (DNMTi) or histone deacetylase inhibitors (HDACi), are approved in monotherapy for cancer treatment. These drugs reprogram gene expression profiles, reactivate tumor suppressor genes (TSG) producing cancer cell differentiation and apoptosis. Epigenetic drugs have been shown to synergize with other epigenetic drugs or various anticancer drugs. To discover new molecular entities that enhance epigenetic therapy, we performed a high-throughput screening using FDA-approved libraries in combination with DNMTi or HDACi. As a screening model, we used YB5 system, a human colon cancer cell line, which contains an epigenetically silenced CMV-GFP locus, mimicking TSG silencing in cancer. CMV-GFP reactivation is triggered by DNMTi or HDACi and responds synergistically to DNMTi/HDACi combination, which phenocopies TSG reactivation upon epigenetic therapy. GFP fluorescence was used as a quantitative readout for epigenetic activity. We discovered that 45 FDA-approved drugs (4% of all drugs tested) in our FDA-approved libraries enhanced DNMTi and HDACi activity, mainly belonging to anticancer and antiarrhythmic drug classes. Transcriptome analysis revealed that combination of decitabine (DNMTi) with the antiarrhythmic proscillaridin A produced profound gene expression reprogramming, which was associated with downregulation of 153 epigenetic regulators, including two known oncogenes in colon cancer (SYMD3 and KDM8). Also, we identified about 85 FDA-approved drugs that antagonized DNMTi and HDACi activity through cytotoxic mechanisms, suggesting detrimental drug interactions for patients undergoing epigenetic therapy. Overall, our drug screening identified new combinations of epigenetic and FDA-approved drugs, which can be rapidly implemented into clinical trials. Mol Cancer Ther; 16(2); 397-407. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/genetics , Drug Repositioning , Epigenesis, Genetic/drug effects , Epigenomics , Gene Expression Regulation, Neoplastic/drug effects , Cell Line, Tumor , Cluster Analysis , Colonic Neoplasms/drug therapy , Computational Biology/methods , DNA Methylation/drug effects , Drug Discovery , Drug Interactions , Drug Screening Assays, Antitumor , Epigenomics/methods , Gene Expression , Gene Expression Profiling , Genes, Reporter , High-Throughput Screening Assays , Histone Deacetylase Inhibitors/pharmacology , Humans , Reproducibility of Results , Small Molecule Libraries
2.
Cancer Res ; 76(6): 1494-505, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26719529

ABSTRACT

Targeting epigenetic pathways is a promising approach for cancer therapy. Here, we report on the unexpected finding that targeting calcium signaling can reverse epigenetic silencing of tumor suppressor genes (TSG). In a screen for drugs that reactivate silenced gene expression in colon cancer cells, we found three classical epigenetic targeted drugs (DNA methylation and histone deacetylase inhibitors) and 11 other drugs that induced methylated and silenced CpG island promoters driving a reporter gene (GFP) as well as endogenous TSGs in multiple cancer cell lines. These newly identified drugs, most prominently cardiac glycosides, did not change DNA methylation locally or histone modifications globally. Instead, all 11 drugs altered calcium signaling and triggered calcium-calmodulin kinase (CamK) activity, leading to MeCP2 nuclear exclusion. Blocking CamK activity abolished gene reactivation and cancer cell killing by these drugs, showing that triggering calcium fluxes is an essential component of their epigenetic mechanism of action. Our data identify calcium signaling as a new pathway that can be targeted to reactivate TSGs in cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Calcium Signaling/drug effects , Calcium/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Epigenesis, Genetic/drug effects , Genes, Tumor Suppressor/drug effects , Calcium Signaling/genetics , Cell Line , Cell Line, Tumor , CpG Islands/drug effects , CpG Islands/genetics , DNA Methylation/drug effects , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Epigenomics/methods , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing/drug effects , HCT116 Cells , HEK293 Cells , HL-60 Cells , Histone Deacetylase Inhibitors/pharmacology , Humans , K562 Cells , Nuclear Proteins/genetics , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
3.
Clin Epigenetics ; 7: 97, 2015.
Article in English | MEDLINE | ID: mdl-26366234

ABSTRACT

BACKGROUND: Aberrant epigenetic silencing of tumor suppressor genes has been recognized as a driving force in cancer. Epigenetic drugs such as the DNA methylation inhibitor decitabine reactivate genes and are effective in myeloid leukemia, but resistance often develops and efficacy in solid tumors is limited. To improve their clinical efficacy, we searched among approved anti-cancer drugs for an epigenetic synergistic combination with decitabine. RESULTS: We used the YB5 cell line, a clonal derivative of the SW48 colon cancer cell line that contains a single copy of a hypermethylated cytomegalovirus (CMV) promoter driving green fluorescent protein (GFP) to screen for drug-induced gene reactivation and synergy with decitabine. None of the 16 anti-cancer drugs tested had effects on their own. However, in combination with decitabine, platinum compounds showed striking synergy in activating GFP. This was dose dependent, observed both in concurrent and sequential combinations, and also seen with other alkylating agents. Clinically achievable concentrations of carboplatin at (25 µM) and decitabine reactivated GFP in 28 % of the YB5 cells as compared to 15 % with decitabine alone. Epigenetic synergy was also seen at endogenously hypermethylated tumor suppressor genes such as MLH1 and PDLIM4. Genome-wide studies showed that reactivation of hypermethylated genes by the combination was significantly better than that induced by decitabine alone or carboplatin alone. Platinum compounds did not enhance decitabine-induced hypomethylation. Rather, we found significantly inhibited HP1α expression by carboplatin and the combination. This was accompanied by increased histone H3 lysine 4 (H3K4) trimethylation and histone H3 lysine 9 (H3K9) acetylation at reactivated genes (P < 0.0001) and reduced occupancy by methyl-binding proteins including MeCP2 and methyl-CpG-binding domain protein 2 (MBD2) (P < 0.0001). CONCLUSIONS: Our results suggest that the combination of decitabine with platinum analogs shows epigenetic synergy that might be exploited in the treatment of different cancers.

4.
Cancer Res ; 72(5): 1170-81, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22219169

ABSTRACT

DNA methylation is commonly thought of as a "molecular lock" that leads to permanent gene silencing. To investigate this notion, we tested 24 different histone deacetylase inhibitors (HDACi) on colon cancer cells that harbor a GFP locus stably integrated and silenced by a hypermethylated cytomegalovirus (CMV) promoter. We found that HDACi efficiently reactivated expression of GFP and many other endogenous genes silenced by DNA hypermethylation. After treatment, all promoters were marked with active chromatin, yet DNA hypermethylation did not change. Thus, DNA methylation could not prevent gene reactivation by drug-induced resetting of the chromatin state. In evaluating the relative contribution of DNA methylation and histone modifications to stable gene silencing, we followed expression levels of GFP and other genes silenced by DNA hypermethylation over time after treatment with HDACi or DNA-demethylating drugs. Reactivation of methylated loci by HDACi was detectable for only 2 weeks, whereas DNA-demethylating drugs induced permanent epigenetic reprogramming. Therefore, DNA methylation cannot be considered as a lock for gene expression but rather as a memory signal for long-term maintenance of gene silencing. These findings define chromatin as an important druggable target for cancer epigenetic therapy and suggest that removal of DNA methylation signals is required to achieve long-term gene reactivation.


Subject(s)
Colonic Neoplasms/genetics , DNA Methylation , Gene Expression Regulation, Neoplastic , Gene Silencing , Cell Line, Tumor , Chromatin Assembly and Disassembly , DNA Methylation/drug effects , Depsipeptides/pharmacology , Green Fluorescent Proteins/genetics , Histone Deacetylase Inhibitors/pharmacology , Humans , Promoter Regions, Genetic , Transcriptional Activation/drug effects
5.
PLoS One ; 6(7): e22110, 2011.
Article in English | MEDLINE | ID: mdl-21760961

ABSTRACT

The epigenetic impact of DNA methylation in chronic myelogenous leukemia (CML) is not completely understood. To elucidate its role we analyzed 120 patients with CML for methylation of promoter-associated CpG islands of 10 genes. Five genes were identified by DNA methylation screening in the K562 cell line and 3 genes in patients with myeloproliferative neoplasms. The CDKN2B gene was selected for its frequent methylation in myeloid malignancies and ABL1 as the target of BCR-ABL translocation. Thirty patients were imatinib-naïve (mostly treated by interferon-alpha before the imatinib era), 30 were imatinib-responsive, 50 were imatinib-resistant, and 10 were imatinib-intolerant. We quantified DNA methylation by bisulfite pyrosequencing. The average number of methylated genes was 4.5 per patient in the chronic phase, increasing significantly to 6.2 in the accelerated and 6.4 in the blastic phase. Higher numbers of methylated genes were also observed in patients resistant or intolerant to imatinib. These patients also showed almost exclusive methylation of a putative transporter OSCP1. Abnormal methylation of a Src suppressor gene PDLIM4 was associated with shortened survival independently of CML stage and imatinib responsiveness. We conclude that aberrant DNA methylation is associated with CML progression and that DNA methylation could be a marker associated with imatinib resistance. Finally, DNA methylation of PDLIM4 may help identify a subset of CML patients that would benefit from treatment with Src/Abl inhibitors.


Subject(s)
DNA Methylation/drug effects , DNA Methylation/genetics , Disease Progression , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Piperazines/pharmacology , Pyrimidines/pharmacology , Adolescent , Adult , Aged , Benzamides , Cell Line, Tumor , Female , Gene Expression Regulation, Leukemic/drug effects , Genes, Neoplasm/genetics , Genome, Human/genetics , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Male , Middle Aged , Multivariate Analysis , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Piperazines/adverse effects , Piperazines/therapeutic use , Pyrimidines/adverse effects , Pyrimidines/therapeutic use , Sequence Analysis, DNA , Survival Analysis , Treatment Outcome , Young Adult
6.
EMBO Rep ; 12(3): 238-43, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21331099

ABSTRACT

Worker and queen bees are genetically indistinguishable. However, queen bees are fertile, larger and have a longer lifespan than their female worker counterparts. Differential feeding of larvae with royal jelly controls this caste switching. There is emerging evidence that the queen-bee phenotype is driven by epigenetic mechanisms. In this study, we show that royal jelly--the secretion produced by the hypopharyngeal and mandibular glands of worker bees--has histone deacetylase inhibitor (HDACi) activity. A fatty acid, (E)-10-hydroxy-2-decenoic acid (10HDA), which accounts for up to 5% of royal jelly, harbours this HDACi activity. Furthermore, 10HDA can reactivate the expression of epigenetically silenced genes in mammalian cells. Thus, the epigenetic regulation of queen-bee development is probably driven, in part, by HDACi activity in royal jelly.


Subject(s)
Bees/physiology , Epigenesis, Genetic , Fatty Acids, Monounsaturated/metabolism , Fatty Acids/metabolism , Histone Deacetylase Inhibitors/metabolism , Insect Hormones/metabolism , Animals , Bees/genetics , Bees/growth & development , Bees/metabolism , DNA Methylation , Fatty Acids, Monounsaturated/chemistry , Female , Hierarchy, Social , Larva/growth & development , Larva/metabolism , Phenotype
7.
PLoS One ; 2(5): e399, 2007 May 02.
Article in English | MEDLINE | ID: mdl-17476321

ABSTRACT

BACKGROUND: Alterations in DNA methylation in cancer include global hypomethylation and gene-specific hypermethylation. It is not clear whether these two epigenetic errors are mechanistically linked or occur independently. This study was performed to determine the relationship between DNA hypomethylation, hypermethylation and microsatellite instability in cancer. METHODOLOGY/PRINCIPAL FINDINGS: We examined 61 cancer cell lines and 60 colorectal carcinomas and their adjacent tissues using LINE-1 bisulfite-PCR as a surrogate for global demethylation. Colorectal carcinomas with sporadic microsatellite instability (MSI), most of which are due to a CpG island methylation phenotype (CIMP) and associated MLH1 promoter methylation, showed in average no difference in LINE-1 methylation between normal adjacent and cancer tissues. Interestingly, some tumor samples in this group showed increase in LINE-1 methylation. In contrast, MSI-showed a significant decrease in LINE-1 methylation between normal adjacent and cancer tissues (P<0.001). Microarray analysis of repetitive element methylation confirmed this observation and showed a high degree of variability in hypomethylation between samples. Additionally, unsupervised hierarchical clustering identified a group of highly hypomethylated tumors, composed mostly of tumors without microsatellite instability. We extended LINE-1 analysis to cancer cell lines from different tissues and found that 50/61 were hypomethylated compared to peripheral blood lymphocytes and normal colon mucosa. Interestingly, these cancer cell lines also exhibited a large variation in demethylation, which was tissue-specific and thus unlikely to be resultant from a stochastic process. CONCLUSION/SIGNIFICANCE: Global hypomethylation is partially reversed in cancers with microsatellite instability and also shows high variability in cancer, which may reflect alternative progression pathways in cancer.


Subject(s)
DNA Methylation , Long Interspersed Nucleotide Elements , Microsatellite Repeats/genetics , Neoplasms/genetics , Base Sequence , DNA Primers , Humans
8.
Cancer Res ; 67(5): 1997-2005, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17332327

ABSTRACT

Gene silencing associated with promoter methylation can inactivate tumor suppressor genes (TSG) in cancer. We identified RIL, a LIM domain gene mapping to 5q31, a region frequently deleted in acute myelogenous leukemia (AML) and myelodysplastic syndrome (MDS), as methylated in 55 of 79 (70%) of cancer cell lines tested. In a variety of primary tumors, we found RIL methylation in 55 of 92 (60%) cases, with highest methylation in AML and colon cancer, and in 30 of 83 (36%) MDS samples, whereas normal tissues showed either absence or substantially lower levels of methylation, which correlates with age. RIL is ubiquitously expressed but silenced in methylated cancers and could be reactivated by the hypomethylating agent 5-aza-2'-deoxycytidine. Restoring RIL expression in colon cancer cells by stable transfection resulted in reduced cell growth and clonogenicity and an approximately 2.0-fold increase in apoptosis following UV exposure. In MDS, RIL methylation is a marker of adverse prognosis independent of chromosome 5 and 7 deletions. Our data suggest that RIL is a good candidate TSG silenced by hypermethylation in cancer.


Subject(s)
Apoptosis/genetics , DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Gene Silencing , Myelodysplastic Syndromes/genetics , Cell Proliferation , Chromosome Mapping , Chromosomes, Human, Pair 5 , CpG Islands , HCT116 Cells , HL-60 Cells , Humans , K562 Cells , LIM Domain Proteins , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/pathology , Nucleic Acid Amplification Techniques , Prognosis , Tumor Cells, Cultured
9.
Cancer ; 109(5): 899-906, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17236224

ABSTRACT

BACKGROUND: Resistance to imatinib is a frequent clinical problem in advanced phase chronic myelogenous leukemia (CML). A Phase II study was performed on low-dose decitabine, a DNA methyltransferase inhibitor, in combination with imatinib in patients with CML in accelerated phase (AP) and myeloid blastic phase (BP). METHODS: Patients received decitabine 15 mg/m(2) intravenously daily, 5 days a week for 2 weeks, and imatinib 600 mg orally daily. Global DNA methylation was measured by long interspersed nucleotide element (LINE) bisulfite/pyrosequencing. RESULTS: Twenty-eight patients were enrolled (25 with imatinib resistance; 18 in AP, 10 in BP). A total of 91 cycles (median, 2.5 cycles per patient) was administered. Complete hematologic responses, partial hematologic responses, and hematologic improvement were observed in 9 (32%), 1 (4%), and 2 (7%) patients. Major and minor cytogenetic responses were observed in 5 (18%) and 3 (11%) patients. The hematologic response rate was higher in patients without BCR-ABL kinase mutations (10 of 19, 53%) than in those with mutations (1 of 7, 14%). Median duration of hematologic response was 18 (range, 4 to 107+) weeks. Myelosuppression was the major adverse effect, with neutropenic fever in 9 patients (32%). LINE methylation decreased from 71.6% +/- 0.9% (mean +/- standard error of the mean) to 60.4% +/- 2.0% on Day 5, 60.5% +/- 1.8% on Day 12, and returned to 68.8% +/- 1.4% at peripheral blood recovery. A decrease in LINE methylation tended to be greater in nonresponders than in responders on Days 5 and 12. CONCLUSIONS: Combination therapy with decitabine and imatinib is well tolerated and active in advanced phase CML without BCR-ABL kinase mutations.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Blast Crisis/drug therapy , Adult , Aged , Azacitidine/administration & dosage , Azacitidine/analogs & derivatives , Benzamides , Blast Crisis/genetics , Blast Crisis/metabolism , DNA Methylation , Decitabine , Female , Genes, abl , Humans , Imatinib Mesylate , Male , Middle Aged , Piperazines/administration & dosage , Polymerase Chain Reaction , Pyrimidines/administration & dosage , Survival Analysis , Treatment Outcome
10.
Blood ; 106(10): 3370-3, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16037387

ABSTRACT

An activating 1849G>T mutation of JAK2 (Janus kinase 2) tyrosine kinase was recently described in chronic myeloproliferative disorders (MPDs). Its role in other hematologic neoplasms is unclear. We developed a quantitative pyrosequencing assay and analyzed 374 samples of hematologic neoplasms. The mutation was frequent in polycythemia vera (PV) (86%) and myelofibrosis (95%) but less prevalent in acute myeloid leukemia (AML) with an antecedent PV or myelofibrosis (5 [36%] of 14 patients). JAK2 mutation was also detected in 3 (19%) of 16 patients with Philadelphia-chromosome (Ph)-negative chronic myelogenous leukemia (CML), 2 (18%) of 11 patients with megakaryocytic AML, 7 (13%) of 52 patients with chronic myelomonocytic leukemia, and 1 (1%) of 68 patients with myelodysplastic syndromes. No mutation was found in Ph(+)CML (99 patients), AML M0-M6 (28 patients), or acute lymphoblastic leukemia (20 patients). We conclude that the JAK2 1849G>T mutation is common in Ph(-) MPD but not critical for transformation to the acute phase of these diseases and that it is generally rare in aggressive leukemias.


Subject(s)
Leukemia, Myeloid/genetics , Point Mutation/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Humans , Janus Kinase 2 , Leukemia, Myeloid/metabolism , Polycythemia Vera/genetics , Polycythemia Vera/metabolism , Primary Myelofibrosis/genetics , Primary Myelofibrosis/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism
11.
J Clin Oncol ; 23(17): 3948-56, 2005 Jun 10.
Article in English | MEDLINE | ID: mdl-15883410

ABSTRACT

PURPOSE: To determine the activity of decitabine, a DNA methylation inhibitor, in imatinib-refractory or intolerant chronic myelogenous leukemia. MATERIALS AND METHODS: Thirty-five patients were enrolled in this phase II study (12 in chronic phase, 17 in accelerated phase, and six in blastic phase). Decitabine was administered at 15 mg/m2 intravenously over 1 hour daily, 5 days a week for 2 weeks. DNA methylation was measured using a LINE1 bisulfite/pyrosequencing assay. RESULTS: Complete hematologic responses were seen in 12 patients (34%) and partial hematologic responses in seven patients (20%), for an overall hematologic response rate of 54% (83% in chronic phase, 41% in accelerated phase, and 34% in blastic phase). Major cytogenetic responses were observed in six patients (17%), and minor cytogenetic responses were seen in 10 patients (29%) for an overall cytogenetic response rate of 46%. Median response duration was 3.5 months (range, 2 to 13+ months). Myelosuppression was the major adverse effect, with neutropenic fever in 28 (23%) of 124 courses of therapy. LINE1 methylation decreased from 71.3% +/- 1.4% (mean +/- standard error of the mean) to 60.7% +/- 1.4% after 1 week, 50.9% +/- 2.4% after 2 weeks, and returned to 66.5% +/- 2.7% at recovery of counts (median, 46 days). LINE1 methylation at the end of week 1 did not correlate with subsequent responses. However, at day 12, the absolute decrease in methylation was 14.5% +/- 3.0% versus 26.8% +/- 2.7% in responders versus nonresponders (P = .007). CONCLUSION: Decitabine induces hypomethylation and has clinical activity in imatinib refractory chronic myelogenous leukemia. We hypothesize that the inverse correlation between hypomethylation 2 weeks after therapy and response is due to a cell death mechanism of response, whereby resistant cells can withstand more hypomethylation.


Subject(s)
Azacitidine/analogs & derivatives , Azacitidine/administration & dosage , Drug Resistance, Neoplasm , Enzyme Inhibitors/administration & dosage , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Neoplasm Recurrence, Local/drug therapy , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Adult , Aged , Antimetabolites, Antineoplastic/therapeutic use , Benzamides , DNA Methylation , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Dose-Response Relationship, Drug , Female , Humans , Imatinib Mesylate , Male , Middle Aged , Protein-Tyrosine Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...