Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
2.
Nat Commun ; 9(1): 1921, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29765031

ABSTRACT

Epigenetics contributes to the pathogenesis of immune-mediated diseases like rheumatoid arthritis (RA). Here we show the first comprehensive epigenomic characterization of RA fibroblast-like synoviocytes (FLS), including histone modifications (H3K27ac, H3K4me1, H3K4me3, H3K36me3, H3K27me3, and H3K9me3), open chromatin, RNA expression and whole-genome DNA methylation. To address complex multidimensional relationship and reveal epigenetic regulation of RA, we perform integrative analyses using a novel unbiased method to identify genomic regions with similar profiles. Epigenomically similar regions exist in RA cells and are associated with active enhancers and promoters and specific transcription factor binding motifs. Differentially marked genes are enriched for immunological and unexpected pathways, with "Huntington's Disease Signaling" identified as particularly prominent. We validate the relevance of this pathway to RA by showing that Huntingtin-interacting protein-1 regulates FLS invasion into matrix. This work establishes a high-resolution epigenomic landscape of RA and demonstrates the potential for integrative analyses to identify unanticipated therapeutic targets.


Subject(s)
Arthritis, Rheumatoid/genetics , Epigenesis, Genetic , Fibroblasts/metabolism , Synoviocytes/metabolism , Adult , Aged , Arthritis, Rheumatoid/metabolism , Chromatin/genetics , Chromatin/metabolism , DNA Methylation , Female , Histone Code , Histones/genetics , Histones/metabolism , Humans , Male , Methylation , Middle Aged , Promoter Regions, Genetic
3.
Clin Cancer Res ; 20(24): 6618-30, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25316818

ABSTRACT

PURPOSE: Low-grade serous ovarian carcinomas (LGSC) are Ras pathway-mutated, TP53 wild-type, and frequently associated with borderline tumors. Patients with LGSCs respond poorly to platinum-based chemotherapy and may benefit from pathway-targeted agents. High-grade serous carcinomas (HGSC) are TP53-mutated and are thought to be rarely associated with borderline tumors. We sought to determine whether borderline histology associated with grade 2 or 3 carcinoma was an indicator of Ras mutation, and we explored the molecular relationship between coexisting invasive and borderline histologies. EXPERIMENTAL DESIGN: We reviewed >1,200 patients and identified 102 serous carcinomas with adjacent borderline regions for analyses, including candidate mutation screening, copy number, and gene expression profiling. RESULTS: We found a similar frequency of low, moderate, and high-grade carcinomas with coexisting borderline histology. BRAF/KRAS alterations were common in LGSC; however, we also found recurrent NRAS mutations. Whereas borderline tumors harbored BRAF/KRAS mutations, NRAS mutations were restricted to carcinomas, representing the first example of a Ras oncogene with an obligatory association with invasive serous cancer. Coexisting borderline and invasive components showed nearly identical genomic profiles. Grade 2 cases with coexisting borderline included tumors with molecular features of LGSC, whereas others were typical of HGSC. However, all grade 3 carcinomas with coexisting borderline histology were molecularly indistinguishable from typical HGSC. CONCLUSION: Our findings suggest that NRAS is an oncogenic driver in serous ovarian tumors. We demonstrate that borderline histology is an unreliable predictor of Ras pathway aberration and underscore an important role for molecular classification in identifying patients that may benefit from targeted agents.


Subject(s)
Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/metabolism , Mutation , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Tumor Suppressor Protein p53/genetics , ras Proteins/genetics , ras Proteins/metabolism , Adult , Aged , Aged, 80 and over , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cluster Analysis , Cohort Studies , Cystadenocarcinoma, Serous/mortality , Cystadenocarcinoma, Serous/pathology , DNA Copy Number Variations , Female , Humans , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Polymorphism, Single Nucleotide , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Signal Transduction , Young Adult
4.
J Mol Diagn ; 16(6): 660-72, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25157968

ABSTRACT

Ongoing cancer genome characterization studies continue to elucidate the spectrum of genomic abnormalities that drive many cancers, and in the clinical arena assessment of the driver genetic alterations in patients is playing an increasingly important diagnostic and/or prognostic role for many cancer types. However, the landscape of genomic abnormalities is still unknown for less common cancers, and the influence of specific genotypes on clinical behavior is often still unclear. To address some of these deficiencies, we developed Profile, a prospective cohort study to obtain genomic information on all patients at a large tertiary care medical center for cancer-related care. We enrolled patients with any cancer diagnosis, and, for each patient (unselected for cancer site or type) we applied mass spectrometric genotyping (OncoMap) of 471 common recurrent mutations in 41 cancer-related genes. We report the results of the first 5000 patients, of which 26% exhibited potentially actionable somatic mutations. These observations indicate the utility of genotyping in advancing the field of precision oncology.


Subject(s)
Genotype , Neoplasms/genetics , Humans , Prospective Studies
5.
J Natl Cancer Inst ; 106(8)2014 Aug.
Article in English | MEDLINE | ID: mdl-25063326

ABSTRACT

Myopericytoma (MPC) is a rare tumor with perivascular proliferation of pluripotent stem-cell-like pericytes. Although indolent, MPC may be locally aggressive with recurrent disease. The pathogenesis and diagnostic biomarkers of MPC are poorly understood. We discovered that 15% of benign MPCs (thyroid, skin; 3 of 20 samples) harbored BRAF(WT/V600E); 33.3% (1 of 3 samples) of BRAF(WT/V600E)-MPCs were multifocal/infiltrative/recurrent. Patient-MPC and primary MPC cells harbored BRAF(WT/V600E), were clonal and expressed pericytic-differentiation biomarkers crucial for its microenvironment. BRAF(WT/V600E)-positive thyroid MPC primary cells triggered in vitro (8.8-fold increase) and in vivo (3.6-fold increase) angiogenesis. Anti-BRAF(V600E) therapy with vemurafenib disrupted angiogenic and metabolic properties (~3-fold decrease) with down-regulation (~2.2-fold decrease) of some extracellular-matrix (ECM) factors and ECM-associated long non-coding RNA (LincRNA) expression, with no effects in BRAF(WT)-pericytes. Vemurafenib also inhibited (~3-fold decrease) cell viability in vitro and in BRAF(WT/V600E)-positive thyroid MPC patient-derived xenograft (PDX) mice (n = 5 mice per group). We established the first BRAF(WT/V600E)-dependent thyroid MPC cell culture. Our findings identify BRAF(WT/V600E) as a novel genetic aberration in MPC pathogenesis and MPC-associated biomarkers and imply that anti-BRAF(V600E) agents may be useful adjuvant therapy in BRAF(WT/V600E)-MPC patients. Patients with BRAF(WT/V600E)-MPC should be closely followed because of the risk for multifocality/recurrence.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Biomarkers, Tumor/genetics , Hemangiopericytoma/pathology , Indoles/pharmacology , Mutation , Pericytes/pathology , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/pharmacology , Thyroid Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Genotype , Glutamic Acid , Hemangiopericytoma/genetics , Humans , Mass Spectrometry , Neoplasm Recurrence, Local/genetics , Thyroid Neoplasms/genetics , Valine , Vemurafenib , Xenograft Model Antitumor Assays
6.
Cancer ; 119(21): 3776-83, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24037752

ABSTRACT

BACKGROUND: Cervical cancer is the second leading cause of cancer deaths among women worldwide. The objective of this study was to describe the most common oncogenic mutations in cervical cancers and to explore genomic differences between the 2 most common histologic subtypes: adenocarcinoma and squamous cell carcinoma. METHODS: A high-throughput genotyping platform, termed Oncomap, was used to interrogate 80 cervical tumors for 1250 known mutations in 139 cancer genes. Samples were analyzed using a mass spectrometry-based genotyping platform and were validated using orthogonal chemistry. Epidermal growth factor receptor (EGFR) mutations were further validated by massive parallel sequencing. Human papilloma virus (HPV) genotyping also was performed. RESULTS: Validated mutations were detected in 48 of 80 tumors (60%) examined. The highest mutation rates were in the genes phosphatidylinositol 3-kinase, catalytic subunit α (PIK3CA) (31.3%); Kirsten rat sarcoma viral oncogene homolog (KRAS) (8.8%); and EGFR (3.8%). PIK3CA mutation rates did not differ significantly between adenocarcinomas and squamous cell carcinomas (25% vs 37.5%, respectively; P = .33). In contrast, KRAS mutations were identified only in adenocarcinomas (17.5% vs 0%; P = .01), and a novel EGFR mutation was detected only in squamous cell carcinomas (0% vs 7.5%; P = .24). There were no associations between HPV-16 or HPV-18 and somatic mutations or overall survival. In adjusted analyses, PIK3CA mutations were associated with shorter survival (67.1 months vs 90.3 months; hazard ratio, 9.1; 95% confidence interval, 2.8-29.5 months; P < .001). CONCLUSIONS: Cervical cancers harbor high rates of potentially targetable oncogenic mutations. In addition, cervical squamous cell carcinoma and adenocarcinoma have distinct molecular profiles, suggesting that clinical outcomes may be improved with the use of more tailored treatment strategies, including PI3K and MEK inhibitors.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Squamous Cell/genetics , Mutation , Oncogenes/genetics , Uterine Cervical Neoplasms/genetics , Adenocarcinoma/epidemiology , Adenocarcinoma/mortality , Adenocarcinoma/virology , Adult , Carcinoma, Squamous Cell/epidemiology , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/virology , Class I Phosphatidylinositol 3-Kinases , Cohort Studies , DNA Mutational Analysis/statistics & numerical data , Female , Gene Frequency , Genes, erbB-1/genetics , Genotype , Humans , Middle Aged , Phosphatidylinositol 3-Kinases/genetics , SEER Program/statistics & numerical data , Uterine Cervical Neoplasms/epidemiology , Uterine Cervical Neoplasms/mortality , Uterine Cervical Neoplasms/virology
7.
PLoS One ; 8(9): e74950, 2013.
Article in English | MEDLINE | ID: mdl-24066160

ABSTRACT

It has been demonstrated for some cancers that the frequency of somatic oncogenic mutations may vary in ancestral populations. To determine whether key driver alterations might occur at different frequencies in colorectal cancer, we applied a high-throughput genotyping platform (OncoMap) to query 385 mutations across 33 known cancer genes in colorectal cancer DNA from 83 Asian, 149 Black and 195 White patients. We found that Asian patients had fewer canonical oncogenic mutations in the genes tested (60% vs Black 79% (P = 0.011) and White 77% (P = 0.015)), and that BRAF mutations occurred at a higher frequency in White patients (17% vs Asian 4% (P = 0.004) and Black 7% (P = 0.014)). These results suggest that the use of genomic approaches to elucidate the different ancestral determinants harbored by patient populations may help to more precisely and effectively treat colorectal cancer.


Subject(s)
Colorectal Neoplasms/genetics , Mutation Rate , Proto-Oncogene Proteins B-raf/genetics , Adult , Aged , Aged, 80 and over , Asian People , Black People , Female , Genotype , Humans , Male , Middle Aged , White People , Young Adult
8.
Nat Genet ; 45(3): 285-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23334667

ABSTRACT

Meningiomas are the most common primary nervous system tumor. The tumor suppressor NF2 is disrupted in approximately half of all meningiomas, but the complete spectrum of genetic changes remains undefined. We performed whole-genome or whole-exome sequencing on 17 meningiomas and focused sequencing on an additional 48 tumors to identify and validate somatic genetic alterations. Most meningiomas had simple genomes, with fewer mutations, rearrangements and copy-number alterations than reported in other tumors in adults. However, several meningiomas harbored more complex patterns of copy-number changes and rearrangements, including one tumor with chromothripsis. We confirmed focal NF2 inactivation in 43% of tumors and found alterations in epigenetic modifiers in an additional 8% of tumors. A subset of meningiomas lacking NF2 alterations harbored recurrent oncogenic mutations in AKT1 (p.Glu17Lys) and SMO (p.Trp535Leu) and exhibited immunohistochemical evidence of activation of these pathways. These mutations were present in therapeutically challenging tumors of the skull base and higher grade. These results begin to define the spectrum of genetic alterations in meningiomas and identify potential therapeutic targets.


Subject(s)
Brain Neoplasms/genetics , Meningioma/genetics , Proto-Oncogene Proteins c-akt/genetics , Receptors, G-Protein-Coupled/genetics , Base Sequence , Brain Neoplasms/pathology , Genomics , Humans , Meningioma/pathology , Mutation , Neurofibromin 2/genetics , Neurofibromin 2/metabolism , Smoothened Receptor
9.
Invest Ophthalmol Vis Sci ; 53(11): 6991-6, 2012 Oct 09.
Article in English | MEDLINE | ID: mdl-22977135

ABSTRACT

PURPOSE: We assessed for mutations in a large number of oncogenes and tumor suppressor genes in primary uveal melanomas using a high-throughput profiling system. METHODS: DNA was extracted and purified from 134 tissue samples from fresh-frozen tissues (n = 87) or formalin-fixed, paraffin-embedded tissues (n = 47) from 124 large uveal melanomas that underwent primary treatment by enucleation. DNA was subjected to whole genome amplification and MALDI-TOF mass spectrometry-based mutation profiling (>1000 mutations tested across 120 oncogenes and tumor suppressor genes) using the OncoMap3 platform. All candidate mutations, as well as commonly occurring mutations in GNAQ and GNA11, were validated using homogeneous mass extension (hME) technology. RESULTS: Of 123 samples, 97 (79%, representing 89 unique tumors) were amplified successfully, passed all quality control steps, and were assayed with the OncoMap platform. A total of 58 mutation calls was made for 49 different mutations across 26 different genes in 34/98 (35%) samples. Of 91 tumors that underwent hME validation, 83 (91%) harbored mutations in the GNAQ (47%) or GNA11 (44%) genes, while hME validation revealed two tumors with mutations in EGFR. These additional mutations occurred in tumors that also had mutations in GNAQ or GNA11. CONCLUSIONS: The vast majority of primary large uveal melanomas harbor mutually-exclusive mutations in GNAQ or GNA11, but very rarely have the oncogenic mutations that are reported commonly in other cancers. When present, these other mutations were found in conjunction with GNAQ/GNA11 mutations, suggesting that these other mutations likely are not the primary drivers of oncogenesis in uveal melanoma.


Subject(s)
GTP-Binding Protein alpha Subunits/genetics , Melanoma/genetics , Mutation/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Uveal Neoplasms/genetics , DNA Mutational Analysis , DNA, Neoplasm/genetics , GTP-Binding Protein alpha Subunits, Gq-G11 , Gene Amplification , Gene Expression Profiling , Humans , Oncogenes/genetics , Tumor Suppressor Proteins/genetics
10.
PLoS One ; 7(8): e41655, 2012.
Article in English | MEDLINE | ID: mdl-22870241

ABSTRACT

BACKGROUND: Given the high incidence of metastatic esophageal squamous cell carcinoma, especially in Asia, we screened for the presence of somatic mutations using OncoMap platform with the aim of defining subsets of patients who may be potential candidate for targeted therapy. METHODS AND MATERIALS: We analyzed 87 tissue specimens obtained from 80 patients who were pathologically confirmed with esophageal squamous cell carcinoma and received 5-fluoropyrimidine/platinum-based chemotherapy. OncoMap 4.0, a mass-spectrometry based assay, was used to interrogate 471 oncogenic mutations in 41 commonly mutated genes. Tumor specimens were prepared from primary cancer sites in 70 patients and from metastatic sites in 17 patients. In order to test the concordance between primary and metastatic sites from the patient for mutations, we analyzed 7 paired (primary-metastatic) specimens. All specimens were formalin-fixed paraffin embedded tissues and tumor content was >70%. RESULTS: In total, we have detected 20 hotspot mutations out of 80 patients screened. The most frequent mutation was PIK3CA mutation (four E545K, five H1047R and one H1047L) (N = 10, 11.5%) followed by MLH1 V384D (N = 7, 8.0%), TP53 (R306, R175H and R273C) (N = 3, 3.5%), BRAF V600E (N = 1, 1.2%), CTNNB1 D32N (N = 1, 1.2%), and EGFR P733L (N = 1, 1.2%). Distributions of somatic mutations were not different according to anatomic sites of esophageal cancer (cervical/upper, mid, lower). In addition, there was no difference in frequency of mutations between primary-metastasis paired samples. CONCLUSIONS: Our study led to the detection of potentially druggable mutations in esophageal SCC which may guide novel therapies in small subsets of esophageal cancer patients.


Subject(s)
Carcinoma, Squamous Cell/genetics , Esophageal Neoplasms/genetics , Mutation, Missense , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins B-raf/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adult , Aged , Amino Acid Substitution , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Class I Phosphatidylinositol 3-Kinases , Disease-Free Survival , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Female , Humans , Male , Middle Aged , MutL Protein Homolog 1 , Neoplasm Metastasis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Pyrimidines/administration & dosage , Survival Rate , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
11.
Eur Urol ; 62(5): 917-22, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22727996

ABSTRACT

BACKGROUND: Metanephric adenoma (MA) of the kidney is a rare, indolent tumor that may be difficult to differentiate from other small renal masses (SRMs). Genetic alterations associated with MA remain largely unknown. OBJECTIVE: We aimed at defining genetic events in MA of the kidney and determining their influence in the management of this disease. DESIGN, SETTING, AND PARTICIPANTS: Multiplexed mass spectrometric genotyping was performed on 29 MA cases after tumor DNA extraction. We also conducted a mutational screen in an additional 129 renal neoplasms. Immunohistochemistry was performed on the MA cases to assess molecular markers of signaling pathway activation. Patients' baseline characteristics, as well as follow-up data, were captured. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: We used descriptive statistics for baseline clinical characteristics and incidence of mutations. The Wilcoxon rank-sum test was used to correlate patient characteristics with mutational status. RESULTS AND LIMITATIONS: We identified the v-raf murine sarcoma viral oncogene homolog B1 (BRAF) V600E mutation in 26 of 29 MA cases. These results were validated in all cases using the commercially available BRAF Pyro Kit (QIAGEN). In contrast, BRAF mutations were rare in the other 129 non-MA renal neoplasms that were screened. We detected a BRAF mutation (V600E) in only one papillary renal cell carcinoma case. In all MA tumors, we documented expression of phosphorylated mitogen-activated protein kinase and phosphorylated extracellular signal-regulated kinase, accompanied by immunoreactivity for p16 (INK4a). All patients were treated with a partial or radical nephrectomy, and after a median follow-up of 26.5 mo, there were no local or distant recurrences. Limitations include the retrospective nature of this study. CONCLUSIONS: BRAF V600E mutations are present in approximately 90% of all MA cases, serving as a potential valuable diagnostic tool in the differential diagnosis of SRMs undergoing a percutaneous biopsy. The presence of BRAF V600E and mitogen-activated protein kinase activation in a largely benign tumor supports the necessity for secondary events (e.g., p16 loss) in BRAF-driven oncogenesis.


Subject(s)
Adenoma/genetics , Kidney Neoplasms/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Adenoma/enzymology , Adenoma/pathology , Adenoma/surgery , Adult , Aged , Cyclin-Dependent Kinase Inhibitor p16/analysis , DNA Mutational Analysis , Female , Genetic Predisposition to Disease , Humans , Immunohistochemistry , Kidney Neoplasms/enzymology , Kidney Neoplasms/pathology , Kidney Neoplasms/surgery , Male , Middle Aged , Mitogen-Activated Protein Kinases/analysis , Nephrectomy , Phenotype , Phosphorylation , Retrospective Studies , Signal Transduction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Time Factors , Treatment Outcome , United States
12.
PLoS One ; 7(6): e38892, 2012.
Article in English | MEDLINE | ID: mdl-22723903

ABSTRACT

BACKGROUND: Gastric cancer is one of the leading cancer types in incidence and mortality, especially in Asia. In order to improve survival, identification of a catalogue of molecular alterations underlying gastric cancer is a critical step for developing and designing genome-directed therapies. METHODOLOGY/PRINCIPAL FINDINGS: The Center for Cancer Genome Discovery (CCGD) at the Dana-Farber Cancer Institute (DFCI) has adapted a high-throughput genotyping platform to determine the mutation status of a large panel of known cancer genes. The mutation detection platform, termed OncoMap v4, interrogates 474 "hotspot" mutations in 41 genes that are relevant for cancer. We performed OncoMap v4 in formalin-fixed paraffin-embedded (FFPE) tissue specimens from 237 gastric adenocarcinomas. Using OncoMap v4, we found that 34 (14.4%) of 237 gastric cancer patients harbored mutations. Among mutations we screened, PIK3CA mutations were the most frequent (5.1%) followed by p53 (4.6%), APC (2.5%), STK11 (2.1%), CTNNB1 (1.7%), and CDKN2A (0.8%). Six samples harbored concomitant somatic mutations. Mutations of CTNNB1 were significantly more frequent in EBV-associated gastric carcinoma (P = 0.046). Our study led to the detection of potentially druggable mutations in gastric cancer which may guide novel therapies in subsets of gastric cancer patients. CONCLUSIONS/SIGNIFICANCE: Using high throughput mutation screening platform, we identified that PIK3CA mutations were the most frequently observed target for gastric adenocarcinoma.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Mutation , Phosphatidylinositol 3-Kinases/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Adult , Aged , Class I Phosphatidylinositol 3-Kinases , Female , Genome-Wide Association Study , Humans , Male , Middle Aged , Mutation Rate , Neoplasm Staging
13.
Nature ; 483(7391): 603-7, 2012 Mar 28.
Article in English | MEDLINE | ID: mdl-22460905

ABSTRACT

The systematic translation of cancer genomic data into knowledge of tumour biology and therapeutic possibilities remains challenging. Such efforts should be greatly aided by robust preclinical model systems that reflect the genomic diversity of human cancers and for which detailed genetic and pharmacological annotation is available. Here we describe the Cancer Cell Line Encyclopedia (CCLE): a compilation of gene expression, chromosomal copy number and massively parallel sequencing data from 947 human cancer cell lines. When coupled with pharmacological profiles for 24 anticancer drugs across 479 of the cell lines, this collection allowed identification of genetic, lineage, and gene-expression-based predictors of drug sensitivity. In addition to known predictors, we found that plasma cell lineage correlated with sensitivity to IGF1 receptor inhibitors; AHR expression was associated with MEK inhibitor efficacy in NRAS-mutant lines; and SLFN11 expression predicted sensitivity to topoisomerase inhibitors. Together, our results indicate that large, annotated cell-line collections may help to enable preclinical stratification schemata for anticancer agents. The generation of genetic predictions of drug response in the preclinical setting and their incorporation into cancer clinical trial design could speed the emergence of 'personalized' therapeutic regimens.


Subject(s)
Databases, Factual , Drug Screening Assays, Antitumor/methods , Encyclopedias as Topic , Models, Biological , Neoplasms/drug therapy , Neoplasms/pathology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Lineage , Chromosomes, Human/genetics , Clinical Trials as Topic/methods , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genes, ras/genetics , Genome, Human/genetics , Genomics , Humans , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Pharmacogenetics , Plasma Cells/cytology , Plasma Cells/drug effects , Plasma Cells/metabolism , Precision Medicine/methods , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Sequence Analysis, DNA , Topoisomerase Inhibitors/pharmacology
14.
J Clin Oncol ; 30(3): 316-21, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22067401

ABSTRACT

PURPOSE: RAF inhibitors are effective against melanomas with BRAF V600E mutations but may induce keratoacanthomas (KAs) and cutaneous squamous cell carcinomas (cSCCs). The potential of these agents to promote secondary malignancies is concerning. We analyzed cSCC and KA lesions for genetic mutations in an attempt to identify an underlying mechanism for their formation. METHODS: Four international centers contributed 237 KA or cSCC tumor samples from patients receiving an RAF inhibitor (either vemurafenib or sorafenib; n = 19) or immunosuppression therapy (n = 53) or tumors that developed spontaneously (n = 165). Each sample was profiled for 396 known somatic mutations across 33 cancer-related genes by using a mass spectrometric-based genotyping platform. RESULTS: Mutations were detected in 16% of tumors (38 of 237), with five tumors harboring two mutations. Mutations in TP53, CDKN2A, HRAS, KRAS, and PIK3CA were previously described in squamous cell tumors. Mutations in MYC, FGFR3, and VHL were identified for the first time. A higher frequency of activating RAS mutations was found in tumors from patients treated with an RAF inhibitor versus populations treated with a non-RAF inhibitor (21.1% v 3.2%; P < .01), although overall mutation rates between treatment groups were similar (RAF inhibitor, 21.1%; immunosuppression, 18.9%; and spontaneous, 17.6%; P = not significant). Tumor histology (KA v cSCC), tumor site (head and neck v other), patient age (≤ 70 v > 70 years), and sex had no significant impact on mutation rate or type. CONCLUSION: Squamous cell tumors from patients treated with an RAF inhibitor have a distinct mutational profile that supports a mechanism of therapy-induced tumorigenesis in RAS-primed cells. Conceivably, cotargeting of MEK together with RAF may reduce or prevent formation of these tumors.


Subject(s)
Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/genetics , Mutation , Protein Kinase Inhibitors/adverse effects , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Benzenesulfonates/adverse effects , Benzenesulfonates/therapeutic use , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Female , Gene Expression Regulation, Neoplastic , Genotype , Humans , Indoles/adverse effects , Indoles/therapeutic use , Male , Mass Spectrometry , Middle Aged , Mitogen-Activated Protein Kinase Kinases/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Pyridines/adverse effects , Pyridines/therapeutic use , Skin Neoplasms/enzymology , Skin Neoplasms/pathology , Sorafenib , Sulfonamides/adverse effects , Sulfonamides/therapeutic use , Vemurafenib
15.
PLoS One ; 6(9): e24433, 2011.
Article in English | MEDLINE | ID: mdl-21931712

ABSTRACT

BACKGROUND: Epithelial ovarian cancer is the most lethal of all gynecologic malignancies, and high grade serous ovarian cancer (HGSC) is the most common subtype of ovarian cancer. The objective of this study was to determine the frequency and types of point somatic mutations in HGSC using a mutation detection protocol called OncoMap that employs mass spectrometric-based genotyping technology. METHODOLOGY/PRINCIPAL FINDINGS: The Center for Cancer Genome Discovery (CCGD) Program at the Dana-Farber Cancer Institute (DFCI) has adapted a high-throughput genotyping platform to determine the mutation status of a large panel of known cancer genes. The mutation detection protocol, termed OncoMap has been expanded to detect more than 1000 mutations in 112 oncogenes in formalin-fixed paraffin-embedded (FFPE) tissue samples. We performed OncoMap on a set of 203 FFPE advanced staged HGSC specimens. We isolated genomic DNA from these samples, and after a battery of quality assurance tests, ran each of these samples on the OncoMap v3 platform. 56% (113/203) tumor samples harbored candidate mutations. Sixty-five samples had single mutations (32%) while the remaining samples had ≥ 2 mutations (24%). 196 candidate mutation calls were made in 50 genes. The most common somatic oncogene mutations were found in EGFR, KRAS, PDGRFα, KIT, and PIK3CA. Other mutations found in additional genes were found at lower frequencies (<3%). CONCLUSIONS/SIGNIFICANCE: Sequenom analysis using OncoMap on DNA extracted from FFPE ovarian cancer samples is feasible and leads to the detection of potentially druggable mutations. Screening HGSC for somatic mutations in oncogenes may lead to additional therapies for this patient population.


Subject(s)
Mutation , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Alleles , Carcinoma, Ovarian Epithelial , Cohort Studies , DNA Mutational Analysis , DNA Primers/genetics , ErbB Receptors/genetics , Female , Genotype , Humans , Mass Spectrometry/methods
16.
J Natl Cancer Inst ; 101(7): 519-32, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19318631

ABSTRACT

BACKGROUND: Overexpression of the fatty acid synthase (FASN) gene has been implicated in prostate carcinogenesis. We sought to directly assess the oncogenic potential of FASN. METHODS: We used immortalized human prostate epithelial cells (iPrECs), androgen receptor-overexpressing iPrECs (AR-iPrEC), and human prostate adenocarcinoma LNCaP cells that stably overexpressed FASN for cell proliferation assays, soft agar assays, and tests of tumor formation in immunodeficient mice. Transgenic mice expressing FASN in the prostate were generated to assess the effects of FASN on prostate histology. Apoptosis was evaluated by Hoechst 33342 staining and by fluorescence-activated cell sorting in iPrEC-FASN cells treated with stimulators of the intrinsic and extrinsic pathways of apoptosis (ie, camptothecin and anti-Fas antibody, respectively) or with a small interfering RNA (siRNA) targeting FASN. FASN expression was compared with the apoptotic index assessed by the terminal deoxynucleotidyltransferase-mediated UTP end-labeling method in 745 human prostate cancer samples by using the least squares means procedure. All statistical tests were two-sided. RESULTS: Forced expression of FASN in iPrECs, AR-iPrECs, and LNCaP cells increased cell proliferation and soft agar growth. iPrECs that expressed both FASN and androgen receptor (AR) formed invasive adenocarcinomas in immunodeficient mice (12 of 14 mice injected formed tumors vs 0 of 14 mice injected with AR-iPrEC expressing empty vector (P < .001, Fisher exact test); however, iPrECs that expressed only FASN did not. Transgenic expression of FASN in mice resulted in prostate intraepithelial neoplasia, the incidence of which increased from 10% in 8- to 16-week-old mice to 44% in mice aged 7 months or more (P = .0028, Fisher exact test), but not in invasive tumors. In LNCaP cells, siRNA-mediated silencing of FASN resulted in apoptosis. FASN overexpression protected iPrECs from apoptosis induced by camptothecin but did not protect iPrECs from Fas receptor-induced apoptosis. In human prostate cancer specimens, FASN expression was inversely associated with the apoptotic rate (mean percentage of apoptotic cells, lowest vs highest quartile of FASN expression: 2.76 vs 1.34, difference = 1.41, 95% confidence interval = 0.45 to 2.39, Ptrend = .0046). CONCLUSIONS: These observations suggest that FASN can act as a prostate cancer oncogene in the presence of AR and that FASN exerts its oncogenic effect by inhibiting the intrinsic pathway of apoptosis.


Subject(s)
Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Apoptosis , Fatty Acid Synthase, Type I/genetics , Oncogenes , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Animals , Apoptosis/genetics , Bromodeoxyuridine/metabolism , Cell Line, Tumor , Disease Models, Animal , Fatty Acid Synthase, Type I/metabolism , Flow Cytometry , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Immunoblotting , Immunohistochemistry , Male , Mice , Mice, Transgenic , Orchiectomy , RNA, Small Interfering/metabolism , Transplantation, Heterologous , Up-Regulation
17.
Lab Invest ; 88(12): 1340-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18838960

ABSTRACT

Fatty acid synthase (FASN), a key metabolic enzyme for liponeogenesis highly expressed in several human cancers, displays oncogenic properties such as resistance to apoptosis and induction of proliferation when overexpressed. To date, no mechanism has been identified to explain the oncogenicity of FASN in prostate cancer. We generated immortalized prostate epithelial cells (iPrECs) overexpressing FASN, and found that (14)C-acetate incorporation into palmitate synthesized de novo by FASN was significantly elevated in immunoprecipitated Wnt-1 when compared to isogenic cells not overexpressing FASN. Overexpression of FASN caused membranous and cytoplasmic beta-catenin protein accumulation and activation, whereas FASN knockdown by short-hairpin RNA resulted in a reduction in the extent of beta-catenin activation. Orthotopic transplantation of iPrECs overexpressing FASN in nude mice resulted in invasive tumors that overexpressed beta-catenin. A strong significant association between FASN and cytoplasmic (stabilized) beta-catenin immunostaining was found in 862 cases of human prostate cancer after computerized subtraction of the membranous beta-catenin signal (P<0.001, Spearman's rho=0.33). We propose that cytoplasmic stabilization of beta-catenin through palmitoylation of Wnt-1 and subsequent activation of the pathway is a potential mechanism of FASN oncogenicity in prostate cancer.


Subject(s)
Cytoplasm/metabolism , Fatty Acid Synthases/metabolism , Palmitic Acid/metabolism , Prostatic Neoplasms/metabolism , Wnt1 Protein/metabolism , beta Catenin/metabolism , Animals , Base Sequence , Cell Line, Transformed , DNA Primers , Humans , Immunohistochemistry , Male , Mice , Mice, Nude , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , RNA Interference
18.
Clin Cancer Res ; 12(8): 2634-9, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16638877

ABSTRACT

PURPOSE: Accumulating evidences show a higher incidence of hepatic neoplasm in HIV/hepatitis C virus (HCV)-coinfected individuals compared with HCV-monoinfected patients. Treatment with HIV-1 protease inhibitors inhibited cancer-promoted angiogenesis in HIV-infected patients affected by Kaposi sarcoma. We aimed to evaluate the antineoplastic potential activities of the protease inhibitor indinavir (Crixivan) in in vitro and in vivo hepatocarcinoma models. EXPERIMENTAL DESIGN: We analyzed effects of indinavir on cell growth and invasiveness in Huh7 and SK-HEP-1 hepatocarcinoma cell lines and on in vivo tumor growth of the same cells in nude mice. Morphologic and molecular analyses on explanted tumors were carried out to evaluate vascularization and apoptosis. RESULTS: We observed a reduced ability to invade an in vitro extracellular matrix for both cell lines treated with indinavir compared with controls (P = 0,001). Moreover, indinavir treatment was able to inhibit matrix metalloproteinase-2 proteolytic activation, whereas there was no effect on cell proliferation. The drug was also able to delay in vivo tumor growth. The inhibition of tumor growth was statistically significant from days 6 to 21 (P = 0.004 and P = 0.003, respectively). Moreover, the drug showed antiangiogenic and proapoptotic actions, as revealed by vessel count and apoptotic index by terminal deoxynucleotide transferase-mediated nick end labeling in explanted tumors. Finally, treatment with indinavir did not block the production of vascular endothelial growth factor in the tumors. CONCLUSION: Indinavir could be helpful to prevent the development of hepatocarcinomas in HIV/HCV-coinfected individuals. In view of the current trend to substitute protease inhibitors with other antiretroviral agents, this information may have clinical implications.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Indinavir/pharmacology , Liver Neoplasms, Experimental/drug therapy , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , HIV Protease Inhibitors/pharmacology , HIV Protease Inhibitors/therapeutic use , Humans , In Situ Nick-End Labeling , Indinavir/therapeutic use , Liver Neoplasms, Experimental/blood supply , Liver Neoplasms, Experimental/pathology , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Nude , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
19.
J Cell Physiol ; 207(1): 232-7, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16331689

ABSTRACT

With the aim to find novel partners of human Cyclin T2a, we performed a two-hybrid screening in yeast using the full-length cDNA of this cyclin as bait, and a human heart cDNA library as preys source. Upon several interesting genes selected, our attention has been focused on the cDNA coding for PKNalpha, a fatty acid- and Rho-activated serine/threonine protein kinase, having a catalytic domain homologous to protein kinase C family. Co-immunoprecipitation and in vitro pull-down assays independently confirmed the interaction between the two proteins. Luciferase assays, performed on NIH3T3 cell extracts after transfection with a MyoD-responsive promoter, pointed out that PKNalpha was able to enhance MyoD-dependent transcription, and that this effect was further increased when cyclin T2a was co-overexpressed. Finally, overexpression of both Cyclin T2a and PKNalpha in C2C12 cells strongly enhanced the expression of myogenic differentiation markers, such as Myogenin and Myosin Heavy Chain, during starvation-induced differentiation. Taken together, our data strengthen the hypothesis that Cyclin T2a plays a role in muscle differentiation, and propose PKNalpha as a novel partner of Cyclin T2a in this process.


Subject(s)
Cell Differentiation , Cyclins/metabolism , Muscle Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Binding Sites/genetics , Binding, Competitive , Cell Line , Cyclin T , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Cyclins/genetics , DNA, Complementary/genetics , Gene Expression/genetics , Humans , Mice , Muscle Cells/cytology , Mutation , MyoD Protein/genetics , MyoD Protein/metabolism , Myogenin/metabolism , Myosin Heavy Chains/metabolism , NIH 3T3 Cells , Plasmids/genetics , Protein Binding , Protein Kinase C , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transfection , Two-Hybrid System Techniques
20.
Clin Cancer Res ; 11(9): 3175-83, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15867210

ABSTRACT

PURPOSE: Employing an in vitro model system of human melanoma progression, we previously reported ferritin light chain (L-ferritin) gene overexpression in the metastatic phenotype. Here, we attempted to characterize the role of ferritin in the biology of human melanoma and in the progression of this disease. EXPERIMENTAL DESIGN: Starting from the LM human metastatic melanoma cell line, we engineered cell clones in which L-ferritin gene expression was down-regulated by the stable expression of a specific antisense construct. These cells were then assayed for their growth capabilities, chemoinvasive properties, and sensitivity to oxidative stress. Additionally, ferritin protein content in primary and metastatic human melanomas was determined by immunohistochemistry. RESULTS: Artificial L-ferritin down-regulation in the LM cells strongly inhibited proliferation and chemoinvasion in vitro and cell growth in vivo. In addition, L-ferritin down-regulated cells displayed enhanced sensitivity to oxidative stress and to apoptosis. Concurrently, immunohistochemical analysis of a human melanoma tissue array revealed that ferritin expression level in metastatic lesions was significantly higher (P < 0.0001) than in primary melanomas. Furthermore, ferritin expression was constantly up-regulated in autologous lymph node melanoma metastases when compared with the respective primary tumors in a cohort of 11 patients. CONCLUSIONS: These data suggest that high ferritin expression can enhance cell growth and improve resistance to oxidative stress in metastatic melanoma cells by interfering with their cellular antioxidant system. The potential significance of these findings deserves to be validated in a clinical setting.


Subject(s)
Ferritins/physiology , Melanoma, Experimental/pathology , Oxidative Stress , Animals , Apoptosis/drug effects , Catalase/metabolism , Cell Line, Tumor , Cell Proliferation , DNA, Antisense/genetics , DNA, Antisense/metabolism , DNA, Complementary/genetics , Dose-Response Relationship, Drug , Down-Regulation , Fatty Acids, Unsaturated/metabolism , Ferritins/analysis , Ferritins/genetics , Gene Expression , Humans , Hydrogen Peroxide , Immunohistochemistry , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Transfection , Transplantation, Heterologous , Up-Regulation , Vitamin E/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...