Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Cancer Res ; 83(13): 2123-2141, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37129912

ABSTRACT

Adrenocortical carcinoma (ACC) is a rare cancer in which tissue-specific differentiation is paradoxically associated with dismal outcomes. The differentiated ACC subtype CIMP-high is prevalent, incurable, and routinely fatal. CIMP-high ACC possess abnormal DNA methylation and frequent ß-catenin-activating mutations. Here, we demonstrated that ACC differentiation is maintained by a balance between nuclear, tissue-specific ß-catenin-containing complexes, and the epigenome. On chromatin, ß-catenin bound master adrenal transcription factor SF1 and hijacked the adrenocortical super-enhancer landscape to maintain differentiation in CIMP-high ACC; off chromatin, ß-catenin bound histone methyltransferase EZH2. SF1/ß-catenin and EZH2/ß-catenin complexes present in normal adrenals persisted through all phases of ACC evolution. Pharmacologic EZH2 inhibition in CIMP-high ACC expelled SF1/ß-catenin from chromatin and favored EZH2/ß-catenin assembly, erasing differentiation and restraining cancer growth in vitro and in vivo. These studies illustrate how tissue-specific programs shape oncogene selection, surreptitiously encoding targetable therapeutic vulnerabilities. SIGNIFICANCE: Oncogenic ß-catenin can use tissue-specific partners to regulate cellular differentiation programs that can be reversed by epigenetic therapies, identifying epigenetic control of differentiation as a viable target for ß-catenin-driven cancers.


Subject(s)
Adrenal Cortex Neoplasms , Adrenocortical Carcinoma , Humans , beta Catenin/genetics , beta Catenin/metabolism , Adrenocortical Carcinoma/genetics , Adrenocortical Carcinoma/metabolism , Adrenocortical Carcinoma/pathology , Adrenal Cortex Neoplasms/genetics , Adrenal Cortex Neoplasms/pathology , Epigenesis, Genetic , Chromatin/genetics
2.
Clin Cancer Res ; 25(11): 3276-3288, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30770352

ABSTRACT

PURPOSE: Adrenocortical carcinoma (ACC) is a rare, aggressive malignancy with few therapies; however, patients with locoregional disease have variable outcomes. The Cancer Genome Atlas project on ACC (ACC-TCGA) identified that cancers of patients with homogeneously rapidly recurrent or fatal disease bear a unique CpG island hypermethylation phenotype, "CIMP-high." We sought to identify a biomarker that faithfully captures this subgroup.Experimental Design: We analyzed ACC-TCGA data to characterize differentially regulated biological processes, and identify a biomarker that is methylated and silenced exclusively in CIMP-high ACC. In an independent cohort of 114 adrenocortical tumors (80 treatment-naive primary ACC, 22 adrenocortical adenomas, and 12 non-naive/nonprimary ACC), we evaluated biomarker methylation by a restriction digest/qPCR-based approach, validated by targeted bisulfite sequencing. We evaluated expression of this biomarker and additional prognostic markers by qPCR. RESULTS: We show that CIMP-high ACC is characterized by upregulation of cell cycle and DNA damage response programs, and identify that hypermethylation and silencing of G0S2 distinguishes this subgroup. We confirmed G0S2 hypermethylation and silencing is exclusive to 40% of ACC, and independently predicts shorter disease-free and overall survival (median 14 and 17 months, respectively). Finally, G0S2 methylation combined with validated molecular markers (BUB1B-PINK1) stratifies ACC into three groups, with uniformly favorable, intermediate, and uniformly dismal outcomes. CONCLUSIONS: G0S2 hypermethylation is a hallmark of rapidly recurrent or fatal ACC, amenable to targeted assessment using routine molecular diagnostics. Assessing G0S2 methylation is straightforward, feasible for clinical decision-making, and will enable the direction of efficacious adjuvant therapies for patients with aggressive ACC.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Adrenal Cortex Neoplasms/pathology , Adrenocortical Carcinoma/genetics , Adrenocortical Carcinoma/pathology , Cell Cycle Proteins/genetics , DNA Methylation , Adrenal Cortex Neoplasms/mortality , Adrenocortical Carcinoma/mortality , Biomarkers, Tumor , Cell Line, Tumor , CpG Islands , Data Mining , Female , Gene Silencing , Genetic Loci , Humans , Male , Neoplasm Grading , Phenotype , Prognosis , Recurrence
3.
Hum Pathol ; 71: 47-54, 2018 01.
Article in English | MEDLINE | ID: mdl-29079178

ABSTRACT

A significant portion of paragangliomas (PGL) and pheochromocytomas (PCC) occur in patients with hereditary PGL/PCC syndromes, including those with germline mutations in succinate dehydrogenase (SDHx) subunit genes. Recently, germline fumarate hydratase (FH) mutations have been identified in a subset of PGL/PCC, and patients with hereditary leiomyomatosis and renal cell carcinoma (HLRCC) may have an increased risk of developing PGL/PCC. SDHB immunohistochemistry (IHC) has previously been shown to be useful for identifying SDHx-deficient PGL/PCC, however, FH IHC has never been explored in these tumors. Thus, we characterized SDHB and FH IHC in a large cohort of PGL/PCC patients (n = 41) at our institution who were evaluated for hereditary PGL/PCC syndromes. Overall, there was strong, positive correlation between germline SDHx subunit gene mutation status and SDHB IHC status (rφ = 0.77; P < .0001), with high corresponding sensitivity, specificity, positive predictive value, and negative predictive value (95.0%, 81.8%, 82.6%, and 94.7%, respectively). Although SDHB loss by IHC was highly correlated with germline SDHx gene mutations, its utility in this population was dependent on clinicopathologic context: while all head and neck PGL patients with SDHB-deficient tumors had germline SDHx gene mutations, only a small subset (25.0%) of PCC patients with SDHB-deficient tumors harbored a germline SDHx gene mutation. Finally, although our cohort contained only one HLRCC patient, their tumor was FH-deficient by IHC, and all other PGL/PCC showed retained FH IHC. Thus, in the appropriate clinical setting, SDHB and FH IHC may be useful for identifying PGL/PCC patients for Medical Genetics evaluation.


Subject(s)
Adrenal Gland Neoplasms/diagnosis , Fumarate Hydratase/biosynthesis , Neoplastic Syndromes, Hereditary/diagnosis , Paraganglioma/diagnosis , Pheochromocytoma/diagnosis , Succinate Dehydrogenase/biosynthesis , Adolescent , Adrenal Gland Neoplasms/genetics , Adult , Aged , Biomarkers, Tumor/analysis , Female , Humans , Immunohistochemistry , Male , Middle Aged , Neoplastic Syndromes, Hereditary/genetics , Paraganglioma/genetics , Pheochromocytoma/pathology , Succinate Dehydrogenase/genetics , Young Adult
5.
J Clin Endocrinol Metab ; 101(3): 999-1007, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26765578

ABSTRACT

CONTEXT: The use of next-generation sequencing has resulted in the identification of recurrent somatic mutations underlying primary aldosteronism (PA). However, significant gaps remain in our understanding of the relationship between tumor aldosterone synthase (CYP11B2) expression and somatic mutation status. OBJECTIVE: The objective of the study was to investigate tumor CYP11B2 expression and somatic aldosterone-driver gene mutation heterogeneity. METHODS: Fifty-one adrenals from 51 PA patients were studied. Immunohistochemistry for CYP11B2 was performed. Aldosterone-producing adenomas with intratumor CYP11B2 heterogeneity were analyzed for mutation status using targeted next-generation sequencing. DNA was isolated from CYP11B2-positive, CYP11B2-negative, and adjacent normal areas from formalin-fixed, paraffin-embedded sections. RESULTS: Of 51 adrenals, seven (14 %) showed distinct heterogeneity in CYP11B2 by immunohistochemistry, including six adenomas with intratumor heterogeneity and one multinodular hyperplastic adrenal with both CYP11B2-positive and -negative nodules. Of the six adrenocortical adenomas with CYP11B2 heterogeneity, three had aldosterone-regulating mutations (CACNA1D p.F747C, KCNJ5 p.L168R, ATP1A1 p.L104R) only in CYP11B2-positive regions, and one had two different mutations localized to two histologically distinct CYP11B2-positive regions (ATP2B3 p.L424_V425del, KCNJ5 p.G151R). Lastly, one adrenal with multiple CYP11B2-expressing nodules showed different mutations in each (CACNA1D p.F747V and ATP1A1 p.L104R), and no mutations were identified in CYP11B2-negative nodule or adjacent normal adrenal. CONCLUSIONS: Adrenal tumors in patients with PA can demonstrate clear heterogeneity in CYP11B2 expression and somatic mutations in driver genes for aldosterone production. These findings suggest that aldosterone-producing adenoma tumorigenesis can occur within preexisting nodules through the acquisition of somatic mutations that drive aldosterone production.


Subject(s)
Adrenocortical Adenoma/chemistry , Adrenocortical Adenoma/genetics , Aldosterone/biosynthesis , Cytochrome P-450 CYP11B2/genetics , Hyperaldosteronism/genetics , Adrenal Glands/pathology , Adrenalectomy , Adrenocortical Adenoma/pathology , DNA/analysis , DNA/chemistry , Gene Expression , Genetic Heterogeneity , Humans , Hyperaldosteronism/pathology , Immunohistochemistry , Mutation , Sequence Analysis, DNA
6.
Oncotarget ; 6(40): 42445-67, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26636651

ABSTRACT

BRAF(V600E) mutation exerts an essential oncogenic function in many tumors, including papillary thyroid carcinoma (PTC). Although BRAF(V600E) inhibitors are available, lack of response has been frequently observed. To study the mechanism underlying intrinsic resistance to the mutant BRAF(V600E) selective inhibitor vemurafenib, we established short-term primary cell cultures of human metastatic/recurrent BRAF(V600E)-PTC, intrathyroidal BRAF(V600E)-PTC, and normal thyroid (NT). We also generated an early intervention model of human BRAF(V600E)-PTC orthotopic mouse. We find that metastatic BRAF(V600E)-PTC cells elicit paracrine-signaling which trigger migration of pericytes, blood endothelial cells and lymphatic endothelial cells as compared to BRAF(WT)-PTC cells, and show a higher rate of invasion. We further show that vemurafenib therapy significantly suppresses these aberrant functions in non-metastatic BRAF(V600E)-PTC cells but lesser in metastatic BRAF(V600E)-PTC cells as compared to vehicle treatment. These results concur with similar folds of down-regulation of tumor microenvironment-associated pro-metastatic molecules, with no effects in BRAF(WT)-PTC and NT cells. Our early intervention preclinical trial shows that vemurafenib delays tumor growth in the orthotopic BRAF(WT/V600E)-PTC mice. Importantly, we identify high copy number gain of MCL1 (chromosome 1q) and loss of CDKN2A (P16, chromosome 9p) in metastatic BRAF(V600E)-PTC cells which are associated with resistance to vemurafenib treatment. Critically, we demonstrate that combined vemurafenib therapy with BCL2/MCL1 inhibitor increases metastatic BRAF(V600E)-PTC cell death and ameliorates response to vemurafenib treatment as compared to single agent treatment. In conclusion, short-term PTC and NT cultures offer a predictive model for evaluating therapeutic response in patients with PTC. Our PTC pre-clinical model suggests that combined targeted therapy might be an important therapeutic strategy for metastatic and refractory BRAF(V600E)-positive PTC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma/genetics , Drug Resistance, Neoplasm/genetics , Gene Dosage , Genes, p16 , Indoles/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Sulfonamides/pharmacology , Thyroid Neoplasms/genetics , Animals , Apoptosis/drug effects , Blotting, Western , Carcinoma, Papillary , Cell Survival/drug effects , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Heterografts , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mice, Inbred NOD , Mice, SCID , Mutation , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neovascularization, Pathologic , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins B-raf/genetics , Thyroid Cancer, Papillary , Transfection , Vemurafenib
7.
Hum Pathol ; 45(7): 1339-47, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24830619

ABSTRACT

Molecular testing for oncogenic gene alterations provides clinically actionable information essential for the optimal management of follicular cell thyroid cancer. We aimed to establish the distribution and frequency of common oncogenic gene mutations and chromosomal rearrangements in a comprehensive set of benign and malignant thyroid lesions. A case-control study was conducted in 413 surgical cases comprising 17 distinct histopathologic categories, 244 malignant, 169 benign, and 304 double-blinded specimens. Seventeen alterations of BRAF, HRAS, KRAS, NRAS, PAX8, and RET genes were evaluated using a single validated technology platform. Following verification of analytical sensitivity, accuracy, and precision in model and surgical specimens, 152 molecular positive results were generated in lesions representing multiple stages of progression and epithelial differentiation as well as rare subtypes of primary, secondary, or recurring tumors. Single mutations were found in 58% of primary malignant lesions and 12% of benign (P < .001). In the blinded validation set, mutation distribution and frequency were distinct across variants of follicular and papillary carcinomas. BRAF or RET-PTC was detected exclusively in malignant lesions but not in follicular carcinomas (P < .001). RAS or PAX8-PPARG were present in 23% of adenomas, and NRAS was found in a single nonneoplastic lesion (P = .0014). These data substantiate the diagnostic utility of molecular testing for oncogenic mutations and validate its performance in a variety of surgical specimens. Standardized and validated multianalyte molecular panels can complement the preoperative and postoperative assessment of thyroid nodules and support a growing number of clinical and translational applications with potential diagnostic, prognostic, or theranostic utility.


Subject(s)
Adenocarcinoma, Follicular/genetics , Adenoma/genetics , Carcinoma, Papillary/genetics , Oncogenes/genetics , Thyroid Neoplasms/genetics , Adenocarcinoma, Follicular/pathology , Adenocarcinoma, Follicular/surgery , Adenoma/pathology , Adenoma/surgery , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma, Papillary/pathology , Carcinoma, Papillary/surgery , Case-Control Studies , Double-Blind Method , Female , Gene Rearrangement , Humans , Male , Middle Aged , Mutation , Prognosis , Thyroid Gland/pathology , Thyroid Neoplasms/pathology , Thyroid Neoplasms/surgery , Young Adult
8.
Cancer Causes Control ; 21(5): 737-43, 2010 May.
Article in English | MEDLINE | ID: mdl-20087645

ABSTRACT

OBJECTIVE OF THE STUDY: Recent publications have reported an association between colon cancer and human papillomaviruses (HPV), suggesting that HPV infection of the colonic mucosa may contribute to the development of colorectal cancer. METHODS: The GP5+/GP6+ PCR reverse line blot method was used for detection of 37 types of human papillomavirus (HPV) in DNA from paraffin-embedded or frozen tissues from patients with colorectal cancer (n = 279) and normal adjacent tissue (n = 30) in three different study populations, including samples from the United States (n = 73), Israel (n = 106) and Spain (n = 100). Additionally, SPF10 PCR was run on all samples (n = 279) and the Innogenetics INNO-LiPA assay was performed on a subset of samples (n = 15). RESULTS: All samples were negative for all types of HPV using both the GP5+/GP6+ PCR reverse line blot method and the SPF10 INNO-LiPA method. CONCLUSIONS: We conclude that HPV types associated with malignant transformation do not meaningfully contribute to adenocarcinoma of the colon.


Subject(s)
Adenocarcinoma/virology , Alphapapillomavirus/isolation & purification , Colorectal Neoplasms/virology , DNA, Viral/isolation & purification , Papillomavirus Infections/complications , Alphapapillomavirus/genetics , Case-Control Studies , Cell Transformation, Viral , Female , Humans , Israel , Male , Polymerase Chain Reaction , Spain , United States
9.
Clin Cancer Res ; 15(2): 668-76, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19147773

ABSTRACT

PURPOSE: Our understanding of adrenocortical carcinoma (ACC) has improved considerably, yet many unanswered questions remain. For instance, can molecular subtypes of ACC be identified? If so, what is their underlying pathogenetic basis and do they possess clinical significance? EXPERIMENTAL DESIGN: We did a whole genome gene expression study of a large cohort of adrenocortical tissues annotated with clinicopathologic data. Using Affymetrix Human Genome U133 Plus 2.0 oligonucleotide arrays, transcriptional profiles were generated for 10 normal adrenal cortices (NC), 22 adrenocortical adenomas (ACA), and 33 ACCs. RESULTS: The overall classification of adrenocortical tumors was recapitulated using principal component analysis of the entire data set. The NC and ACA cohorts showed little intragroup variation, whereas the ACC cohort revealed much greater variation in gene expression. A robust list of 2,875 differentially expressed genes in ACC compared with both NC and ACA was generated and used in functional enrichment analysis to find pathways and attributes of biological significance. Cluster analysis of the ACCs revealed two subtypes that reflected tumor proliferation, as measured by mitotic counts and cell cycle genes. Kaplan-Meier analysis of these ACC clusters showed a significant difference in survival (P < 0.020). Multivariate Cox modeling using stage, mitotic rate, and gene expression data as measured by the first principal component for ACC samples showed that gene expression data contains significant independent prognostic information (P < 0.017). CONCLUSIONS: This study lays the foundation for the molecular classification and prognostication of adrenocortical tumors and also provides a rich source of potential diagnostic and prognostic markers.


Subject(s)
Adrenal Cortex Neoplasms/diagnosis , Adrenal Cortex Neoplasms/genetics , Gene Expression Profiling , Adrenal Cortex/pathology , Cell Line, Tumor , Cluster Analysis , Cohort Studies , Cyclin E/metabolism , Genome , Humans , Immunohistochemistry/methods , Oligonucleotide Array Sequence Analysis , Prognosis , Proportional Hazards Models , Transcription, Genetic
10.
Clin Cancer Res ; 12(7 Pt 1): 1983-93, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16609007

ABSTRACT

A subset of follicular thyroid carcinomas contains a balanced translocation, t(2;3)(q13;p25), that results in fusion of the paired box gene 8 (PAX8) and peroxisome proliferator-activated receptor gamma (PPARG) genes with concomitant expression of a PAX8-PPARgamma fusion protein, PPFP. PPFP is thought to contribute to neoplasia through a mechanism in which it acts as a dominant-negative inhibitor of wild-type PPARgamma. To better understand this type of follicular carcinoma, we generated global gene expression profiles using DNA microarrays of a cohort of follicular carcinomas along with other common thyroid tumors and used the data to derive a gene expression profile characteristic of PPFP-positive tumors. Transient transfection assays using promoters of four genes whose expression was highly associated with the translocation showed that each can be activated by PPFP. PPFP had unique transcriptional activities when compared with PAX8 or PPARgamma, although it had the potential to function in ways qualitatively similar to PAX8 or PPARgamma depending on the promoter and cellular environment. Bioinformatics analyses revealed that genes with increased expression in PPFP-positive follicular carcinomas include known PPAR target genes; genes involved in fatty acid, amino acid, and carbohydrate metabolism; micro-RNA target genes; and genes on chromosome 3p. These results have implications for the neoplastic mechanism of these follicular carcinomas.


Subject(s)
Adenocarcinoma, Follicular/genetics , Gene Expression Profiling , Oncogene Proteins, Fusion/genetics , PPAR gamma/genetics , Paired Box Transcription Factors/genetics , Thyroid Neoplasms/genetics , Translocation, Genetic , Adenocarcinoma, Follicular/metabolism , Adenocarcinoma, Follicular/pathology , Chromosomes, Human, Pair 2/genetics , Chromosomes, Human, Pair 3/genetics , Computational Biology , Humans , Oncogene Proteins, Fusion/biosynthesis , PAX8 Transcription Factor , PPAR gamma/metabolism , Paired Box Transcription Factors/metabolism , Principal Component Analysis , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology
11.
Oncogene ; 24(44): 6646-56, 2005 Oct 06.
Article in English | MEDLINE | ID: mdl-16007166

ABSTRACT

Thyroid cancer poses a significant clinical challenge, and our understanding of its pathogenesis is incomplete. To gain insight into the pathogenesis of papillary thyroid carcinoma, transcriptional profiles of four normal thyroids and 51 papillary carcinomas (PCs) were generated using DNA microarrays. The tumors were genotyped for their common activating mutations: BRAF V600E point mutation, RET/PTC1 and 3 rearrangement and point mutations of KRAS, HRAS and NRAS. Principal component analysis based on the entire expression data set separated the PCs into three groups that were found to reflect tumor morphology and mutational status. By combining expression profiles with mutational status, we defined distinct expression profiles for the BRAF, RET/PTC and RAS mutation groups. Using small numbers of genes, a simple classifier was able to classify correctly the mutational status of all 40 tumors with known mutations. One tumor without a detectable mutation was predicted by the classifier to have a RET/PTC rearrangement and was shown to contain one by fluorescence in situ hybridization analysis. Among the mutation-specific expression signatures were genes whose differential expression was a direct consequence of the mutation, as well as genes involved in a variety of biological processes including immune response and signal transduction. Expression of one mutation-specific differentially expressed gene, TPO, was validated at the protein level using immunohistochemistry and tissue arrays containing an independent set of tumors. The results demonstrate that mutational status is the primary determinant of gene expression variation within these tumors, a finding that may have clinical and diagnostic significance and predicts success for therapies designed to prevent the consequences of these mutations.


Subject(s)
Gene Expression Profiling , Genes, ras , Mutation , Oligonucleotide Array Sequence Analysis , Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Cell Surface/genetics , Thyroid Neoplasms/genetics , Base Sequence , DNA Primers , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Iodide Peroxidase/metabolism , Proto-Oncogene Proteins c-ret , Receptors, G-Protein-Coupled , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...