Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Mol Cancer Ther ; 15(12): 3087-3096, 2016 12.
Article in English | MEDLINE | ID: mdl-27638859

ABSTRACT

Identification of synthetically lethal cellular targets and synergistic drug combinations is important in cancer chemotherapy as they help to overcome treatment resistance and increase efficacy. The Ataxia Telangiectasia Mutated (ATM) kinase is a nuclear protein that plays a major role in the initiation of DNA repair signaling and cell-cycle check points during DNA damage. Although ATM was shown to be associated with poor prognosis in gastric cancer, its implications as a predictive biomarker for cancer chemotherapy remain unexplored. The present study evaluated ATM-induced synthetic lethality and its role in sensitization of gastric cancer cells to PARP and TOP1 inhibitors, veliparib (ABT-888) and irinotecan (CPT-11), respectively. ATM expression was detected in a panel of gastric cell lines, and the IC50 against each inhibitors was determined. The combinatorial effect of ABT-888 and CPT-11 in gastric cancer cells was also determined both in vitro and in vivo ATM deficiency was found to be associated with enhanced sensitivity to ABT-888 and CPT-11 monotherapy, hence suggesting a mechanism of synthetic lethality. Cells with high ATM expression showed reduced sensitivity to monotherapy; however, they showed a higher therapeutic effect with ABT-888 and CPT-11 combinatorial therapy. Furthermore, ATM expression was shown to play a major role in cellular homeostasis by regulating cell-cycle progression and apoptosis in a P53-independent manner. The present study highlights the clinical utility of ATM expression as a predictive marker for sensitivity of gastric cancer cells to PARP and TOP1 inhibition and provides a deeper mechanistic insight into ATM-dependent regulation of cellular processes. Mol Cancer Ther; 15(12); 3087-96. ©2016 AACR.


Subject(s)
Apoptosis , Ataxia Telangiectasia Mutated Proteins/genetics , Benzimidazoles/pharmacology , Camptothecin/analogs & derivatives , Cell Cycle , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Camptothecin/pharmacology , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Drug Synergism , Gene Expression , Histones/metabolism , Humans , Inhibitory Concentration 50 , Irinotecan , Oncogene Protein p21(ras)/genetics , Oncogene Protein p21(ras)/metabolism , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Xenograft Model Antitumor Assays
2.
Cell Oncol (Dordr) ; 39(2): 175-86, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26868260

ABSTRACT

BACKGROUND: Melanoma-associated antigen (MAGE)-A3 is a member of the family of cancer-testis antigens and has been found to be epigenetically regulated and aberrantly expressed in various cancer types. It has also been found that MAGE-A3 expression may correlate with an aggressive clinical course and with chemo-resistance. The objectives of this study were to assess the relationship between MAGE-A3 promoter methylation and expression and (1) gastric cancer patient survival and (2) its functional consequences in gastric cancer-derived cells. METHODS: Samples from two independent gastric cancer cohorts (including matched non-malignant gastric samples) were included in this study. MAGE-A3 methylation and mRNA expression levels were determined by methylation-specific PCR (MSP) and quantitative real-time PCR (qPCR), respectively. MAGE-A3 expression was knocked down in MKN1 gastric cancer-derived cells using miRNAs. In addition, in vitro cell proliferation, colony formation, apoptosis, cell cycle, drug treatment, immunohistochemistry and Western blot assays were performed. RESULTS: Clinical analysis of 223 primary patient-derived samples (ntumor = 161, nnormal = 62) showed a significant inverse correlation between MAGE-A3 promoter methylation and expression in the cancer samples (R = -0.63, p = 5.99e-19). A lower MAGE-A3 methylation level was found to be associated with a worse patient survival (HR: 1.5, 95 % CI: 1.02-2.37, p = 0.04). In addition, we found that miRNA-mediated knockdown of MAGE-A3 expression in MKN1 cells caused a reduction in its proliferation and colony forming capacities, respectively. Under stress conditions MAGE-A3 was found to regulate the expression of Bax and p21. MAGE-A3 knock down also led to an increase in Puma and Noxa expression, thus contributing to an enhanced docetaxel sensitivity in the gastric cancer-derived cells. CONCLUSIONS: From our results we conclude that MAGE-A3 expression is regulated epigenetically by promoter methylation, and that its expression contributes to gastric cell proliferation and drug sensitivity. This study underscores the potential implications of MAGE-A3 as a therapeutic target and prognostic marker in gastric cancer patients.


Subject(s)
Antigens, Neoplasm/metabolism , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Neoplasm Proteins/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Antigens, Neoplasm/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Methylation/drug effects , DNA Methylation/genetics , Docetaxel , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing/drug effects , Humans , Neoplasm Proteins/genetics , Promoter Regions, Genetic , Stress, Physiological/drug effects , Survival Analysis , Taxoids/pharmacology , Tumor Stem Cell Assay
3.
Exp Hematol ; 43(1): 32-43.e1-35, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25450514

ABSTRACT

The use of genome-wide copy-number analysis and massive parallel sequencing has revolutionized the understanding of the clonal architecture of pediatric acute lymphoblastic leukemia (ALL) by demonstrating that this disease is composed of highly variable clonal ancestries following the rules of Darwinian selection. The current study aimed to analyze the molecular composition of childhood ALL biopsies and patient-derived xenografts with particular emphasis on mechanisms associated with acquired chemoresistance. Genomic DNA from seven primary pediatric ALL patient samples, 29 serially passaged xenografts, and six in vivo selected chemoresistant xenografts were analyzed with 250K single-nucleotide polymorphism arrays. Copy-number analysis of non-drug-selected xenografts confirmed a highly variable molecular pattern of variegated subclones. Whereas primary patient samples from initial diagnosis displayed a mean of 5.7 copy-number alterations per sample, serially passaged xenografts contained a mean of 8.2 and chemoresistant xenografts a mean of 10.5 copy-number alterations per sample, respectively. Resistance to cytarabine was explained by a new homozygous deletion of the DCK gene, whereas methotrexate resistance was associated with monoallelic deletion of FPGS and mutation of the remaining allele. This study demonstrates that selecting for chemoresistance in xenografted human ALL cells can reveal novel mechanisms associated with drug resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Clone Cells/pathology , Neoplastic Stem Cells/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Animals , Antineoplastic Agents/therapeutic use , Biopsy , Cytarabine/pharmacology , Cytarabine/therapeutic use , DNA, Neoplasm/genetics , Deoxycytidine Kinase/genetics , Dexamethasone/therapeutic use , Disease Progression , Drug Resistance, Neoplasm/genetics , Female , Gene Dosage , Heterografts , Humans , Male , Methotrexate/pharmacology , Methotrexate/therapeutic use , Mice , Mice, Inbred NOD , Mice, SCID , Mutation , Neoplasm Proteins/genetics , Neoplasm Transplantation , Peptide Synthases/genetics , Polymorphism, Single Nucleotide , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Radiation Chimera , Sequence Analysis, DNA , Vincristine/therapeutic use
4.
BMC Genomics ; 15 Suppl 9: S20, 2014.
Article in English | MEDLINE | ID: mdl-25521664

ABSTRACT

BACKGROUND: Non-small cell lung cancer (NSCLC) is a major cause of cancer-related death worldwide due to poor patient prognosis and clinical outcome. Here, we studied the genetic variations underlying NSCLC pathogenesis based on their association to patient outcome after gemcitabine therapy. RESULTS: Bioinformatics analysis was used to investigate possible effects of POLA2 G583R (POLA2+1747 GG/GA, dbSNP ID: rs487989) in terms of protein function. Using biostatistics, POLA2+1747 GG/GA (rs487989, POLA2 G583R) was identified as strongly associated with mortality rate and survival time among NSCLC patients. It was also shown that POLA2+1747 GG/GA is functionally significant for protein localization via green fluorescent protein (GFP)-tagging and confocal laser scanning microscopy analysis. The single nucleotide polymorphism (SNP) causes DNA polymerase alpha subunit B to localize in the cytoplasm instead of the nucleus. This inhibits DNA replication in cancer cells and confers a protective effect in individuals with this SNP. CONCLUSIONS: The results suggest that POLA2+1747 GG/GA may be used as a prognostic biomarker of patient outcome in NSCLC pathogenesis.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Computational Biology , DNA Polymerase I/genetics , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Polymorphism, Single Nucleotide , Active Transport, Cell Nucleus , Adult , Aged , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Nucleus/metabolism , DNA Polymerase I/chemistry , DNA Polymerase I/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Female , Genotype , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/drug therapy , Male , Middle Aged , Models, Molecular , Mutation , Prognosis , Protein Conformation , Survival Analysis , Gemcitabine
5.
BMC Gastroenterol ; 14: 55, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24674026

ABSTRACT

BACKGROUND: Methylation-induced silencing of promoter CpG islands in tumor suppressor genes plays an important role in human carcinogenesis. In colorectal cancer, the CpG island methylator phenotype (CIMP) is defined as widespread and elevated levels of DNA methylation and CIMP+ tumors have distinctive clinicopathological and molecular features. In contrast, the existence of a comparable CIMP subtype in gastric cancer (GC) has not been clearly established. To further investigate this issue, in the present study we performed comprehensive DNA methylation profiling of a well-characterised series of primary GC. METHODS: The methylation status of 1,421 autosomal CpG sites located within 768 cancer-related genes was investigated using the Illumina GoldenGate Methylation Panel I assay on DNA extracted from 60 gastric tumors and matched tumor-adjacent gastric tissue pairs. Methylation data was analysed using a recursively partitioned mixture model and investigated for associations with clinicopathological and molecular features including age, Helicobacter pylori status, tumor site, patient survival, microsatellite instability and BRAF and KRAS mutations. RESULTS: A total of 147 genes were differentially methylated between tumor and matched tumor-adjacent gastric tissue, with HOXA5 and hedgehog signalling being the top-ranked gene and signalling pathway, respectively. Unsupervised clustering of methylation data revealed the existence of 6 subgroups under two main clusters, referred to as L (low methylation; 28% of cases) and H (high methylation; 72%). Female patients were over-represented in the H tumor group compared to L group (36% vs 6%; P = 0.024), however no other significant differences in clinicopathological or molecular features were apparent. CpG sites that were hypermethylated in group H were more frequently located in CpG islands and marked for polycomb occupancy. CONCLUSIONS: High-throughput methylation analysis implicates genes involved in embryonic development and hedgehog signaling in gastric tumorigenesis. GC is comprised of two major methylation subtypes, with the highly methylated group showing some features consistent with a CpG island methylator phenotype.


Subject(s)
Adenocarcinoma/genetics , CpG Islands , DNA Methylation , Gene Expression Regulation, Neoplastic/genetics , Stomach Neoplasms/genetics , Adenocarcinoma/complications , Adenocarcinoma/metabolism , Age Factors , Aged , Case-Control Studies , Cyclin A1/genetics , Female , Helicobacter Infections/complications , Helicobacter pylori , Homeodomain Proteins/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/genetics , Microsatellite Instability , Middle Aged , Phenotype , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras) , Sex Factors , Signal Transduction , Stomach Neoplasms/complications , Stomach Neoplasms/metabolism , ras Proteins/genetics
6.
PLoS One ; 9(3): e91666, 2014.
Article in English | MEDLINE | ID: mdl-24626295

ABSTRACT

We aimed to identify a prostate cancer DNA hypermethylation microarray signature (denoted as PHYMA) that differentiates prostate cancer from benign prostate hyperplasia (BPH), high from low-grade and lethal from non-lethal cancers. This is a non-randomized retrospective study in 111 local Asian men (87 prostate cancers and 24 BPH) treated from 1995 to 2009 in our institution. Archival prostate epithelia were laser-capture microdissected and genomic DNA extracted and bisulfite-converted. Samples were profiled using Illumina GoldenGate Methylation microarray, with raw data processed by GenomeStudio. A classification model was generated using support vector machine, consisting of a 55-probe DNA methylation signature of 46 genes. The model was independently validated on an internal testing dataset which yielded cancer detection sensitivity and specificity of 95.3% and 100% respectively, with overall accuracy of 96.4%. Second validation on another independent western cohort yielded 89.8% sensitivity and 66.7% specificity, with overall accuracy of 88.7%. A PHYMA score was developed for each sample based on the state of methylation in the PHYMA signature. Increasing PHYMA score was significantly associated with higher Gleason score and Gleason primary grade. Men with higher PHYMA scores have poorer survival on univariate (p = 0.0038, HR = 3.89) and multivariate analyses when controlled for (i) clinical stage (p = 0.055, HR = 2.57), and (ii) clinical stage and Gleason score (p = 0.043, HR = 2.61). We further performed bisulfite genomic sequencing on 2 relatively unknown genes to demonstrate robustness of the assay results. PHYMA is thus a signature with high sensitivity and specificity for discriminating tumors from BPH, and has a potential role in early detection and in predicting survival.


Subject(s)
DNA Methylation , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , Aged , Aged, 80 and over , Asian People , Cell Differentiation , Epigenesis, Genetic , Gene Expression Profiling , Humans , Male , Middle Aged , Multivariate Analysis , Oligonucleotide Array Sequence Analysis , Prognosis , Proportional Hazards Models , Prostatic Hyperplasia/diagnosis , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/ethnology , Reproducibility of Results , Retrospective Studies , Sensitivity and Specificity
7.
Mol Carcinog ; 53(2): 98-108, 2014 Feb.
Article in English | MEDLINE | ID: mdl-22911899

ABSTRACT

The degree of gene hypermethylation in non-neoplastic colonic mucosa (NNCM) is a potentially important event in the development of colorectal cancer (CRC), particularly for the subgroup with a CpG island methylator phenotype (CIMP). In this study, we aimed to use an unbiased and high-throughput approach to evaluate the topography of DNA methylation in the non-neoplastic colonic mucosa (NNCM) surrounding colorectal cancer (CRC). A total of 61 tissue samples comprising 53 NNCM and 8 tumor samples were obtained from hemicolectomy specimens of two CRC patients (Cases 1 and 2). NNCM was stripped from the underlying colonic wall and samples taken at varying distances from the tumor. The level of DNA methylation in NNCM and tumor tissues was assessed at 1,505 CpG sites in 807 cancer-related genes using Illumina GoldenGate® methylation arrays. Case 1 tumor showed significantly higher levels of methylation compared to surrounding NNCM samples (P < 0.001). The average level of methylation in NNCM decreased with increasing distance from the tumor (r = -0.418; P = 0.017), however this was not continuous and "patches" with higher levels of methylation were observed. Case 2 tumor was less methylated than Case 1 tumor (average ß-value 0.181 vs. 0.415) and no significant difference in the level of methylation was observed in comparison to the surrounding NNCM. No evidence was found for a diminishing gradient of methylation in the NNCM surrounding CRC with a high level of methylation. Further work is required to determine whether CIMP+ CRC develop from within "patches" of NCCM that display high levels of methylation.


Subject(s)
Colon/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Methylation , Intestinal Mucosa/pathology , Aged , CpG Islands , Female , Humans , Male , Middle Aged
8.
Gut ; 62(1): 22-33, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22535375

ABSTRACT

OBJECTIVE: Cisplatin is a widely used gastric cancer (GC) chemotherapy; however, genetic factors regulating GC responses to cisplatin remain obscure. Identifying genes regulating cisplatin resistance could aid clinicians in tailoring treatments, by distinguishing cisplatin sensitive patients from those who might benefit from alternative platinum therapies, and highlight novel targeted strategies for overcoming cisplatin resistance. Here integrated epigenomics is applied to identify genes associated with GC cisplatin resistance. DESIGN: 20 GC cell lines were subjected to gene expression profiling, DNA methylation profiling and drug response assays. The molecular data were integrated to identify genes highly expressed and unmethylated specifically in cisplatin-resistant lines. Candidate genes were functionally tested by several in vitro and in vivo assays. Clinical impact of candidate genes was also assessed in a cohort of 197 GC patients. RESULTS: Epigenomic analysis identified bone morphogenetic protein 4 (BMP4) as an epigenetically regulated gene highly expressed in cisplatin-resistant lines. Functional assays confirmed that BMP4 is necessary and sufficient for the expression of several prooncogenic traits, likely mediated through stimulation of the epithelial-mesenchymal transition. In primary tumours, BMP4 promoter methylation levels were inversely correlated with BMP4 expression, and patients with high BMP4-expressing tumours exhibited significantly worse prognosis. Therapeutically, targeted genetic inhibition of BMP4 caused significant sensitisation of GC cells to cisplatin. Notably, BMP4-expressing GCs also did not exhibit cross resistance to oxaliplatin. CONCLUSIONS: BMP4 epigenetic and expression status may represent promising biomarkers for GC cisplatin resistance. Targeting BMP4 may sensitise GC cells to cisplatin. Oxaliplatin, a clinically acceptable cisplatin alternative, may represent a potential therapeutic option for BMP4-positive GCs.


Subject(s)
Adenocarcinoma/genetics , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Bone Morphogenetic Protein 4/genetics , Cisplatin/pharmacology , Drug Resistance, Neoplasm/genetics , Stomach Neoplasms/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Bone Morphogenetic Protein 4/metabolism , Cell Line, Tumor , Cisplatin/therapeutic use , DNA Methylation/drug effects , Epigenomics/methods , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Humans , Organoplatinum Compounds/pharmacology , Organoplatinum Compounds/therapeutic use , Oxaliplatin , Prognosis , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality
9.
PLoS One ; 7(12): e50786, 2012.
Article in English | MEDLINE | ID: mdl-23226540

ABSTRACT

Although gemcitabine is highly active in several cancer types, intrinsic and acquired drug resistance remains a major challenge. Overexpression of Bcl-2 has been associated with gemcitabine resistance. The aim of this study is to determine whether gossypol can overcome gemcitabine resistance in cell lines with high level of Bcl-2 expression in combination drug therapy. Our study demonstrated that in 10 cell lines derived from different cancers, high Bcl-2 baseline expression was observed in cell lines that were resistant to gemcitabine (GEM-R). Furthermore, synergistic effect of combination therapy was observed in gemcitabine-resistant (GEM-R) cell lines with high Bcl-2 expression, but not in a gemcitabine-sensitive (GEM-S) cell lines regardless of Bcl-2 expression. Gossypol treatment resulted in the decrease of anti-apoptotic genes such as Bcl-2 and Bcl-xl and an upregulation of the pro-apoptotic gene, Noxa. Furthermore, the addition of gossypol to gemcitabine resulted in lower expressions of anti-apoptotic genes compared to gemcitabine alone. Gene expression profiling in GEM-R and GEM-S cell lines suggest that anti-apoptotic genes such as pAkt and PI3KR2 may play important role in gemcitabine resistance, while pro-apoptotic Bcl-2 related genes (Bad, Caspase-6 and Calpain-1) may regulate synergistic interaction in combination therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm , Gossypol/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Down-Regulation/drug effects , Down-Regulation/genetics , Drug Interactions , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Gossypol/analogs & derivatives , Gossypol/pharmacology , Humans , Inhibitory Concentration 50 , Multigene Family/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Reproducibility of Results , Signal Transduction/drug effects , Signal Transduction/genetics , Up-Regulation/drug effects , Up-Regulation/genetics , bcl-X Protein/genetics , bcl-X Protein/metabolism , Gemcitabine
10.
Sci Transl Med ; 4(156): 156ra140, 2012 Oct 17.
Article in English | MEDLINE | ID: mdl-23076357

ABSTRACT

Epigenetic alterations are fundamental hallmarks of cancer genomes. We surveyed the landscape of DNA methylation alterations in gastric cancer by analyzing genome-wide CG dinucleotide (CpG) methylation profiles of 240 gastric cancers (203 tumors and 37 cell lines) and 94 matched normal gastric tissues. Cancer-specific epigenetic alterations were observed in 44% of CpGs, comprising both tumor hyper- and hypomethylation. Twenty-five percent of the methylation alterations were significantly associated with changes in tumor gene expression. Whereas most methylation-expression correlations were negative, several positively correlated methylation-expression interactions were also observed, associated with CpG sites exhibiting atypical transcription start site distances and gene body localization. Methylation clustering of the tumors revealed a CpG island methylator phenotype (CIMP) subgroup associated with widespread hypermethylation, young patient age, and adverse patient outcome in a disease stage-independent manner. CIMP cell lines displayed sensitivity to 5-aza-2'-deoxycytidine, a clinically approved demethylating drug. We also identified long-range regions of epigenetic silencing (LRESs) in CIMP tumors. Combined analysis of the methylation, gene expression, and drug treatment data suggests that certain LRESs may silence specific genes within the region, rather than all genes. Finally, we discovered regions of long-range tumor hypomethylation, associated with increased chromosomal instability. Our results provide insights into the epigenetic impact of environmental and biological agents on gastric epithelial cells, which may contribute to cancer.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Stomach Neoplasms/genetics , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Proliferation , Chromosomal Instability , Cluster Analysis , CpG Islands , Decitabine , Epigenomics , Gastric Mucosa/metabolism , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Mice , Neoplasm Transplantation , Phenotype
11.
PLoS One ; 7(3): e33894, 2012.
Article in English | MEDLINE | ID: mdl-22479469

ABSTRACT

Current regimens for induction therapy of pediatric acute lymphoblastic leukemia (ALL), or for re-induction post relapse, use a combination of vincristine (VCR), a glucocorticoid, and L-asparaginase (ASP) with or without an anthracycline. With cure rates now approximately 80%, robust pre-clinical models are necessary to prioritize active new drugs for clinical trials in relapsed/refractory patients, and the ability of these models to predict synergy/antagonism with established therapy is an essential attribute. In this study, we report optimization of an induction-type regimen by combining VCR, dexamethasone (DEX) and ASP (VXL) against ALL xenograft models established from patient biopsies in immune-deficient mice. We demonstrate that the VXL combination was synergistic in vitro against leukemia cell lines as well as in vivo against ALL xenografts. In vivo, VXL treatment caused delays in progression of individual xenografts ranging from 22 to >146 days. The median progression delay of xenografts derived from long-term surviving patients was 2-fold greater than that of xenografts derived from patients who died of their disease. Pharmacokinetic analysis revealed that systemic DEX exposure in mice increased 2-fold when administered in combination with VCR and ASP, consistent with clinical findings, which may contribute to the observed synergy between the 3 drugs. Finally, as proof-of-principle we tested the in vivo efficacy of combining VXL with either the Bcl-2/Bcl-xL/Bcl-w inhibitor, ABT-737, or arsenic trioxide to provide evidence of a robust in vivo platform to prioritize new drugs for clinical trials in children with relapsed/refractory ALL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Induction Chemotherapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Asparaginase/administration & dosage , Biphenyl Compounds/administration & dosage , Cell Line, Tumor , Child , Dexamethasone/administration & dosage , Disease Models, Animal , Drug Synergism , Female , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Nitrophenols/administration & dosage , Piperazines/administration & dosage , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Sulfonamides/administration & dosage , Vincristine/administration & dosage , Xenograft Model Antitumor Assays
12.
Diagn Mol Pathol ; 19(4): 243-7, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21051995

ABSTRACT

The recent emergence of high-throughput arrays for methylation analysis has made the influence of tumor content on the interpretation of methylation levels increasingly pertinent. However, to what degree does tumor content have an influence, and what degree of tumor content makes a specimen acceptable for accurate analysis remains unclear. Taking a systematic approach, we analyzed 98 unselected formalin-fixed and paraffin-embedded gastric tumors and matched normal tissue samples using the Illumina GoldenGate methylation assay. Unsupervised hierarchical clustering showed 2 separate clusters with a significant difference in average tumor content levels. The probes identified to be significantly differentially methylated between the tumors and normals also differed according to the tumor content of the samples included, with the sensitivity of identifying the "top" candidate probes significantly reduced when including samples below 70% tumor content. We also tested whether the removal of the probes featuring single nucleotide polymorphisms and/or DNA repetitive elements, reportedly present in GoldenGate arrays, would significantly affect the study's findings, and found little change in the results with their omission. Our findings suggest that tumor content significantly influences the interpretation of methylation levels and candidate gene identification, and that 70% tumor content may be a suitable threshold for selecting samples for methylation studies.


Subject(s)
DNA/chemistry , Pathology, Molecular/methods , Stomach Neoplasms/diagnosis , Stomach Neoplasms/pathology , Cluster Analysis , DNA Methylation , Humans , Paraffin Embedding , Sensitivity and Specificity
13.
BMC Cancer ; 10: 227, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20492682

ABSTRACT

BACKGROUND: Most previous studies of the CpG island methylator phenotype (CIMP) in colorectal cancer (CRC) have been conducted on a relatively small numbers of CpG sites. In the present study we performed comprehensive DNA methylation profiling of CRC with the aim of characterizing CIMP subgroups. METHODS: DNA methylation at 1,505 CpG sites in 807 cancer-related genes was evaluated using the Illumina GoldenGate methylation array in 28 normal colonic mucosa and 91 consecutive CRC samples. Methylation data was analyzed using unsupervised hierarchical clustering. CIMP subgroups were compared for various clinicopathological and molecular features including patient age, tumor site, microsatellite instability (MSI), methylation at a consensus panel of CpG islands and mutations in BRAF and KRAS. RESULTS: A total of 202 CpG sites were differentially methylated between tumor and normal tissue. Unsupervised hierarchical clustering of methylation data from these sites revealed the existence of three CRC subgroups referred to as CIMP-low (CIMP-L, 21% of cases), CIMP-mid (CIMP-M, 14%) and CIMP-high (CIMP-H, 65%). In comparison to CIMP-L tumors, CIMP-H tumors were more often located in the proximal colon and showed more frequent mutation of KRAS and BRAF (P<0.001). CONCLUSIONS: Comprehensive DNA methylation profiling identified three CRC subgroups with distinctive clinicopathological and molecular features. This study suggests that both KRAS and BRAF mutations are involved with the CIMP-H pathway of CRC rather than with distinct CIMP subgroups.


Subject(s)
Colorectal Neoplasms/genetics , CpG Islands , DNA Methylation , Gene Expression Profiling , Adult , Age Factors , Aged , Cluster Analysis , Colorectal Neoplasms/pathology , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Male , Microsatellite Instability , Middle Aged , Mutation , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras) , ras Proteins/genetics
14.
J Pharmacol Exp Ther ; 324(2): 434-42, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17986648

ABSTRACT

The microtubule-depolymerizing drug, vincristine, is effective in the treatment of acute lymphoblastic leukemia (ALL). Although vincristine resistance mechanisms have been extensively characterized in cell lines, their clinical relevance is poorly understood. The aim of the current study was to define clinically relevant mechanisms of vincristine resistance in a panel of childhood ALL xenografts established in immune-deficient (nonobese diabetic/severe combined immunodeficient) mice. We also studied two independent xenograft sublines that were selected by in vivo vincristine exposure. In vitro vincristine sensitivity determined by a stromal coculture, murine bone marrow stromal cell line (MS-5), assay, but not methyl-thiazolyl-tetrazolium metabolic activity assay, significantly correlated (P = 0.05) with the length of the patients' first remission. Investigations into mechanisms of resistance revealed no association with steady-state vincristine accumulation or increased activity and/or expression of ATP-binding cassette transporters, although increased intracellular levels of polymerized tubulin significantly correlated with resistance (r = 0.85; P = 0.0019). Two xenograft sublines selected by in vivo vincristine exposure exhibited a 2-fold increase in polymerized tubulin levels compared with the parental subline (P < 0.05), reflecting their in vivo vincristine resistance. In this study, a vincristine-resistant xenograft with high levels of polymerized tubulin was relatively sensitive to the microtubule-polymerizing drug paclitaxel. These results indicate that the balance between polymerized and nonpolymerized tubulin may be an important determinant of response to Vinca alkaloid-based chemotherapy regimens in childhood ALL.


Subject(s)
Drug Resistance, Neoplasm/physiology , Microtubules/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Vincristine/therapeutic use , Xenograft Model Antitumor Assays/methods , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Humans , Mice , Microtubules/drug effects , Polymers/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Vincristine/pharmacology
15.
Proteomics ; 6(5): 1681-94, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16456880

ABSTRACT

Intrinsic or acquired resistance to vincristine (VCR), an antimicrotubule agent used in the treatment of childhood acute lymphoblastic leukemia (ALL), is a major clinical problem. Using a clinically relevant NOD/SCID mouse xenograft model of ALL, we established that alterations in the actin and tubulin cytoskeleton are involved in in vivo VCR resistance. Altered protein expression between VCR-sensitive ALL xenografts, and xenografts with intrinsic or acquired VCR resistance, was identified using 2-D DIGE coupled with MS. Of the 19 proteins displaying altered expression, 11 are associated with the actin cytoskeleton. Altered expression of the actin- and/or tubulin-binding proteins gelsolin, moesin, ezrin, tropomyosin, CAP-G, HSP27, HSP70, TCP-1, and stathmin were associated with in vivo VCR resistance. The actin-regulating protein gelsolin was increased in both acquired and resistant leukemia as confirmed by immunoblotting and gene expression. The major cytoskeletal protein, gamma-actin, was down-regulated in the VCR-resistant leukemia xenografts; in contrast, there was no significant change in beta-actin expression. This study provides the first evidence for a role of the actin cytoskeleton in intrinsic and acquired in vivo antimicrotubule drug resistance in childhood leukemia and highlights the power of 2-D DIGE for the discovery of resistance markers, pharmacoproteomics, and signaling pathways in cancer.


Subject(s)
Actins/metabolism , Antineoplastic Agents, Phytogenic/therapeutic use , Cytoskeleton/metabolism , Drug Resistance, Neoplasm , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Proteome/analysis , Actins/chemistry , Animals , Child , Cytoskeleton/chemistry , Electrophoresis, Gel, Two-Dimensional , Humans , Mice , Mice, SCID , Microtubule Proteins/chemistry , Microtubule Proteins/metabolism , Molecular Sequence Data , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transplantation, Heterologous , Vincristine
16.
Methods Mol Med ; 111: 323-34, 2005.
Article in English | MEDLINE | ID: mdl-15911988

ABSTRACT

Acute lymphoblastic leukemia (ALL) is predominantly a disease of the bone marrow that disseminates to multiple organ sites throughout the body and, without aggressive treatment, eventually results in multiorgan failure and death. Experimental models that mimic the dissemination of ALL have been difficult to establish, principally due to the poor engraftment efficiency of normal and malignant human hematopoietic cells in various strains of immune-deficient mice. The recent availability of mouse strains that are even more immunocompromised than established strains such as the nude (nu/nu) or severe combined immunodeficient (SCID) mouse has presented opportunities to establish improved experimental models of human leukemia. In this chapter we outline the methodology to (1) establish continuous xenografts from primary childhood ALL biopsies in nonobese diabetic/SCID (NOD/SCID) mice and (2) utilize these xenograft models of systemic disease to test established and experimental drugs while monitoring leukemia progression in "real time" by serial monitoring of murine peripheral blood. These experimental models will be useful for the preclinical evaluation of novel therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Animals , Biopsy , Disease Progression , Humans , In Vitro Techniques , Mice , Mice, Nude , Mice, SCID , Neoplasm Transplantation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
17.
Blood ; 103(10): 3905-14, 2004 May 15.
Article in English | MEDLINE | ID: mdl-14764536

ABSTRACT

Continuous xenografts from 10 children with acute lymphoblastic leukemia (ALL) were established in nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice. Relative to primary engrafted cells, negligible changes in growth rates and immunophenotype were observed at second and third passage. Analysis of clonal antigen receptor gene rearrangements in 2 xenografts from patients at diagnosis showed that the pattern of clonal variation observed following tertiary transplantation in mice exactly reflected that in bone marrow samples at the time of clinical relapse. Patients experienced diverse treatment outcomes, including 5 who died of disease (median, 13 months; range, 11-76 months, from date of diagnosis), and 5 who remain alive (median, 103 months; range, 56-131 months, following diagnosis). When stratified according to patient outcome, the in vivo sensitivity of xenografts to vincristine and dexamethasone, but not methotrexate, differed significantly (P =.028, P =.029, and P =.56, respectively). The in vitro sensitivity of xenografts to dexamethasone, but not vincristine, correlated significantly with in vivo responses and patient outcome. This study shows, for the first time, that the biologic and genetic characteristics, and patterns of chemosensitivity, of childhood ALL xenografts accurately reflect the clinical disease. As such, they provide powerful experimental models to prioritize new therapeutic strategies for future clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Disease Models, Animal , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Transplantation, Heterologous , Adolescent , Animals , Antineoplastic Agents/therapeutic use , Child , Child, Preschool , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Drug Evaluation, Preclinical , Female , Gene Rearrangement, T-Lymphocyte , Genes, Immunoglobulin , Humans , Immunophenotyping , Male , Methotrexate/pharmacology , Methotrexate/therapeutic use , Mice , Mice, SCID , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Treatment Outcome , Vincristine/pharmacology , Vincristine/therapeutic use
18.
Blood ; 99(11): 4100-8, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12010813

ABSTRACT

Acute lymphoblastic leukemia cells from 19 children, including 7 who remain in first complete remission (CR1), were engrafted into nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice. High-level infiltration of bone marrow, spleen, and liver was observed, with variable infiltration of other organs. The immunophenotypes of xenografts were essentially unaltered compared with the original patient sample. In addition, sequencing of the entire p53 coding region revealed no mutations in 14 of 14 xenografts (10 from patients at diagnosis and 4 at relapse). Cells harvested from the spleens of engrafted mice readily transferred the leukemia to secondary and tertiary recipients. To correlate biologic characteristics of xenografts with clinical and prognostic features of the patients, the rates at which individual leukemia samples engrafted in NOD/SCID mice were analyzed. Differences in biologic correlates were encountered depending on stage of disease: a direct correlation was observed between the rate of engraftment and length of CR1 for samples harvested at relapse (r = 0.96; P =.002), but not diagnosis (r = 0.38; P =.40). In contrast, the in vivo responses of 6 xenografts to vincristine showed a direct correlation (r = 0.96; P =.002) between the length of CR1 and the rate at which the leukemia cell population recovered following vincristine treatment, regardless of whether the xenografts were derived from patients at diagnosis or relapse. This study supports previous findings that the NOD/SCID model of childhood ALL provides an accurate representation of the human disease and indicates that it may be of value to predict relapse and design alternative treatment strategies in a patient-specific manner.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma/physiopathology , Adolescent , Animals , B-Lymphocytes/pathology , Child , Child, Preschool , Disease Models, Animal , Disease-Free Survival , Female , Humans , Kinetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Recurrence , T-Lymphocytes/pathology , Time Factors , Transplantation, Heterologous , Tumor Cells, Cultured , Vincristine/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...