Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
2.
PLoS One ; 12(4): e0176139, 2017.
Article in English | MEDLINE | ID: mdl-28445481

ABSTRACT

Colorectal cancer (CRC) is the second leading cause of cancer death in the United States. In the metastatic setting, the majority of patients respond to initial therapies but eventually develop resistance and progress. In this study, we test the hypothesis that priming with epigenetic therapy sensitizes CRC cell lines, which were previously resistant to subsequent chemotherapeutic agents. When multiple CRC cell lines are first exposed to 500 nM of the DNA demethylating agent, 5-aza-cytidine (AZA) in-vitro, and the cells then established as in-vivo xenografts in untreated NOD-SCID mice; there is an enhanced response to cytotoxic chemotherapy with agents commonly used in CRC treatment. For irinotecan (IRI), growth diminished by 16-62 fold as assessed, by both proliferation (IC50) and anchorage independent cell growth soft agar assays. Treatment of resistant HCT116 cell line along with in-vivo, for CRC line xenografts, AZA plus IRI again exhibits this synergistic response with significant improvement in survival and tumor regression in the mice. Genome-wide expression correlates changes in pathways for cell adhesion and DNA repair with the above responses. A Phase 1/2 clinical trial testing this concept is already underway testing the clinical efficacy of this concept in IRI resistant, metastatic CRC (NCT01896856).


Subject(s)
Antineoplastic Agents/therapeutic use , Azacitidine/therapeutic use , Camptothecin/analogs & derivatives , Colorectal Neoplasms/drug therapy , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Antineoplastic Agents/toxicity , Azacitidine/toxicity , Caco-2 Cells , Camptothecin/therapeutic use , Camptothecin/toxicity , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA Methylation/drug effects , DNA Repair/drug effects , Gene Expression/drug effects , Gene Expression Profiling , HCT116 Cells , Humans , Irinotecan , Long Interspersed Nucleotide Elements/genetics , Mice , Mice, Inbred NOD , Mice, SCID
3.
Epigenetics ; 12(7): 540-550, 2017 07 03.
Article in English | MEDLINE | ID: mdl-27918237

ABSTRACT

Epigenetic control of gene expression is a major determinant of tumor phenotype and has been found to influence sensitivity to individual chemotherapeutic agents. Glutathione peroxidase 3 (GPX3, plasma glutathione peroxidase) is a key component of cellular antioxidant regulation and its gene has been reported to be methylated in specific tumor types. GPX3 role in oxidative damage has been associated with sensitivity to platinums in other tumors but its importance in colorectal cancer (CRC) has not been determined. We examined the role of GPX3 methylation in colorectal carcinoma in determining sensitivity to platinum drugs using primary tumor specimens, cell lines, knockdown cell lines, and tumor cell line xenografts. We find GPX3 promoter region methylation in approximately one third of CRC samples and GPX3 methylation leads to reduced GPX3 expression and increased oxaliplatin and cisplatin sensitivity. In contrast, in cell lines with high baseline levels of GPX3 expression or with the ability to increase GPX3 expression, platinum resistance is increased. The cisplatin IC50 in GPX3-methylated cell lines is approximately 6-fold lower than that in GPX3-unmethylated lines. Additionally, knockdown cell lines with essentially no GPX3 expression require N-acetylcysteine to survive in culture underscoring the importance of GPX3 in redox biology. In vivo, GPX3 methylation predicts tumor xenograft sensitivity to platinum with regression of GPX3 knockdown xenografts with platinum treatment but continued growth of GPX3 wild type xenografts in the presence of platinum. These studies demonstrate the importance of GPX3 for CRC cells resistance to platinums and the potential utility of GPX3 methylation status as a predictive biomarker for platinum sensitivity in CRC.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma/genetics , Colorectal Neoplasms/genetics , DNA Methylation , Drug Resistance, Neoplasm/genetics , Glutathione Peroxidase/genetics , Promoter Regions, Genetic , Animals , Antineoplastic Agents/therapeutic use , Caco-2 Cells , Carcinoma/drug therapy , Carcinoma/pathology , Cisplatin/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Female , Glutathione Peroxidase/metabolism , HCT116 Cells , HT29 Cells , Humans , Mice , Mice, Nude , Organoplatinum Compounds/therapeutic use , Oxaliplatin
4.
BMC Med Inform Decis Mak ; 15: 84, 2015 Oct 14.
Article in English | MEDLINE | ID: mdl-26467091

ABSTRACT

BACKGROUND: Pediatric asthma affects 7.1 million American children incurring an annual total direct healthcare cost around 9.3 billion dollars. Asthma control in children is suboptimal, leading to frequent asthma exacerbations, excess costs, and decreased quality of life. Successful prediction of risk for asthma control deterioration at the individual patient level would enhance self-management and enable early interventions to reduce asthma exacerbations. We developed and tested the first set of models for predicting a child's asthma control deterioration one week prior to occurrence. METHODS: We previously reported validation of the Asthma Symptom Tracker, a weekly asthma self-monitoring tool. Over a period of two years, we used this tool to collect a total of 2912 weekly assessments of asthma control on 210 children. We combined the asthma control data set with patient attributes and environmental variables to develop machine learning models to predict a child's asthma control deterioration one week ahead. RESULTS: Our best model achieved an accuracy of 71.8 %, a sensitivity of 73.8 %, a specificity of 71.4 %, and an area under the receiver operating characteristic curve of 0.757. We also identified potential improvements to our models to stimulate future research on this topic. CONCLUSIONS: Our best model successfully predicted a child's asthma control level one week ahead. With adequate accuracy, the model could be integrated into electronic asthma self-monitoring systems to provide real-time decision support and personalized early warnings of potential asthma control deteriorations.


Subject(s)
Asthma/diagnosis , Models, Statistical , Adolescent , Child , Child, Preschool , Female , Humans , Machine Learning , Male , Prognosis , Sensitivity and Specificity
5.
Am J Pathol ; 184(1): 260-70, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24200853

ABSTRACT

Large-magnitude numerical distinctions (>10-fold) among drug responses of genetically contrasting cancers were crucial for guiding the development of some targeted therapies. Similar strategies brought epidemiological clues and prevention goals for genetic diseases. Such numerical guides, however, were incomplete or low magnitude for Fanconi anemia pathway (FANC) gene mutations relevant to cancer in FANC-mutation carriers (heterozygotes). We generated a four-gene FANC-null cancer panel, including the engineering of new PALB2/FANCN-null cancer cells by homologous recombination. A characteristic matching of FANCC-null, FANCG-null, BRCA2/FANCD1-null, and PALB2/FANCN-null phenotypes was confirmed by uniform tumor regression on single-dose cross-linker therapy in mice and by shared chemical hypersensitivities to various inter-strand cross-linking agents and γ-radiation in vitro. Some compounds, however, had contrasting magnitudes of sensitivity; a strikingly high (19- to 22-fold) hypersensitivity was seen among PALB2-null and BRCA2-null cells for the ethanol metabolite, acetaldehyde, associated with widespread chromosomal breakage at a concentration not producing breaks in parental cells. Because FANC-defective cancer cells can share or differ in their chemical sensitivities, patterns of selective hypersensitivity hold implications for the evolutionary understanding of this pathway. Clinical decisions for cancer-relevant prevention and management of FANC-mutation carriers could be modified by expanded studies of high-magnitude sensitivities.


Subject(s)
Acetaldehyde/pharmacology , Drug Resistance, Neoplasm/genetics , Fanconi Anemia Complementation Group Proteins/genetics , Animals , Blotting, Western , Cell Line, Tumor , Fanconi Anemia/genetics , Humans , Mice , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays
6.
Int J Cancer ; 134(3): 596-605, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-23873170

ABSTRACT

Phenotypic differences among cancers with the same origin may be associated with chemotherapy response. CHFR silencing associated with DNA methylation has been suggested to be predictive of taxane sensitivity in diverse tumor types. However, the use of microsatellite instability (MSI:unstable-MSS:stable) as a predictive marker for therapeutic effect has had conflicting results. We examined these molecular alterations as predictors of chemotherapy sensitivity in colorectal cancer (CRC). Differential sensitivity to docetaxel and gemcitabine was compared to potential predictive biomarkers CHFR methylation and MSI status. Cell lines that were MSI-H/CHFR-methylated, MSS/CHFR-methylated and MSS/CHFR-unmethylated were assessed for in vivo sensitivity of CRC cell line xenografts to docetaxel and/or gemcitabine. We observed increased sensitivity in vitro to gemcitabine in cell lines with MSI and docetaxel in cell lines with CHFR inactivation via DNA methylation. In vivo treatment of human xenografts confirmed differential sensitivity, with the MSI-H/CHFR-methylated line RKO having tumor growth inhibition to each agent, and at least additive tumor growth inhibition with combination therapy. The MSS-CHFR-unmethylated line, CACO2 , was resistant to single and combination therapy, while COLO205, the MSS/CHFR-methylated line, showed tumor growth inhibition with docetaxel, but not gemcitabine, therapy. CHFR methylation in CRC cell lines predicted for sensitivity in vitro and in vivo to docetaxel, while MSI-H cell lines were more sensitive to gemcitabine. These data suggest that a subset of CRC patients would be selectively sensitive to a novel combination of gemcitabine and docetaxel, and are the basis for an ongoing clinical trial of this combination in a biomarker-selected patient population.


Subject(s)
Antineoplastic Agents/therapeutic use , Cell Cycle Proteins/genetics , Colorectal Neoplasms/drug therapy , Deoxycytidine/analogs & derivatives , Gene Silencing , Microsatellite Instability , Neoplasm Proteins/genetics , Taxoids/therapeutic use , Animals , Base Sequence , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Methylation , DNA Primers , Deoxycytidine/therapeutic use , Docetaxel , Female , Humans , Mice , Poly-ADP-Ribose Binding Proteins , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction , Ubiquitin-Protein Ligases , Xenograft Model Antitumor Assays , Gemcitabine
7.
Oncotarget ; 4(2): 298-309, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23593653

ABSTRACT

Epigenetic inactivation of tumor-suppressor and other regulatory genes plays a critical role in carcinogenesis. Transcriptional silencing is often maintained by DNA methyl transferase (DNMT)-mediated hypermethylation of CpG islands in promoter DNA. Nucleoside analogs including azacytidine and decitabine have been used to inhibit DNMT and re-activate genes, and are clinically used. Their shortcomings include a short half-life and a slow onset of action due to required nucleotide incorporation during DNA replication, which may limit clinical utility. It might be useful to begin to identify lead compounds having novel properties, specifically distinct and fast-acting gene desilencing. We previously identified chemicals augmenting gene expression in multiple reporter systems. We now report that a subset of these compounds that includes quinacrine re-expresses epigenetically silenced genes implicated in carcinogenesis. p16, TFPI2, the cadherins E-cadherin and CDH13, and the secreted frizzle-related proteins (SFRPs) SFRP1 and SFRP5 were desilenced in cancer cell lines. These lead compounds were fast-acting: re-expression occurred by 12-24 hours. Reactivation of silenced genes was accompanied by depletion of DNMT1 at the promoters of activated genes and demethylation of DNA. A model compound, 5175328, induced changes more rapidly than decitabine. These gene desilencing agents belonged to a class of acridine compounds, intercalated into DNA, and inhibited DNMT1 activity in vitro. Although to define the mechanism would be outside the scope of this initial report, this class may re-activate silenced genes in part by intercalating into DNA and subsequently inhibiting full DNMT1 activity. Rapid mechanisms for chemical desilencing of methylated genes therefore exist.


Subject(s)
DNA Methylation/drug effects , Gene Silencing/drug effects , Genes, Tumor Suppressor/drug effects , Intercalating Agents/pharmacology , Acridines/pharmacology , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , DNA/drug effects , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/metabolism , Epigenomics , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans
8.
Oncotarget ; 3(7): 678-85, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22824673

ABSTRACT

5-Fluorouracil (5FU) and similar fluoropyrimidines induce covalent modification of thymidylate synthase (TS) and inhibit its activity. They are often used to treat solid cancers, but drug resistance and toxicity are drawbacks. Therefore, there is an unmet need for a functional assay to quantify fluorouracil activity in tissues, so as to individually tailor dosing. It is cumbersome to separately quantify unmodified and 5FU-modified TS using currently available commercial anti-TS antibodies because they recognize both forms. We report here the first monoclonal antibody (FTS) specific to 5FU-modified TS. By immunoblot assay, the FTS antibody specifically recognizes modified TS in a dose-dependent manner in 5FU-treated cells, in cancer xenograft tissues of 5FU-treated mice, and in the murine tissues. In the same assay, the antibody is nonreactive with unmodified TS in untreated or treated cells and tissues. Speculatively, a high-throughput assay could be enabled by pairing anti-TS antibodies of two specificities, one recognizing only modified TS and another recognizing both forms, to structurally quantify the TS-inhibiting effect of fluorouracil at a cellular or tissue level without requiring prior protein separation. Such a development might aid preclinical analytic studies or make practical the individual tailoring of dosing.


Subject(s)
Antibodies, Monoclonal/chemistry , Fluorouracil/analysis , Fluorouracil/immunology , Thymidylate Synthase/analysis , Thymidylate Synthase/immunology , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Colorectal Neoplasms/chemistry , Colorectal Neoplasms/drug therapy , Dose-Response Relationship, Drug , Drug Monitoring/methods , Female , Floxuridine/analysis , Floxuridine/immunology , Floxuridine/pharmacology , Fluorouracil/pharmacology , Immunoenzyme Techniques/methods , Methotrexate/analysis , Methotrexate/pharmacology , Mice , Mice, Nude , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/drug therapy , Rats , Thymidylate Synthase/antagonists & inhibitors , Transplantation, Heterologous , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...