Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Urol Oncol ; 33(3): 108.e5-13, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25175425

ABSTRACT

OBJECTIVE: Androgen-induced E26 transformation-specific (ETS) gene fusion-positive tumors have been associated with aggressive prostate cancer. The aim is to evaluate the ETS gene rearrangement status on initial biopsy of patients registered in the Reduction by Dutasteride of Clinical Progression Events in Expectant Management trial study and determine if gene fusion status was associated with disease progression. MATERIALS AND METHODS: Initial biopsy material from 146 men registered in Reduction by Dutasteride of Clinical Progression Events in Expectant Management trial study treated with dutasteride (73/146, 50%) and as placebo (73/146, 50%) were reviewed, and ERG and SPINK1 immunohistochemistry was performed. ERG- and SPINK1-negative cancer samples were evaluated for ETV1, ETV4, and ETV5 rearrangements by fluorescence in situ hybridization. Frequency of ETS gene aberrations in both groups was correlated with cancer progression including prostate-specific antigen progression, Gleason progression, and progression-free survival by logistic analysis, pairwise differences, and chunk likelihood ratio tests for the genotype groups. RESULTS AND CONCLUSIONS: Of the 146 patients, 99 (67.8%) (placebo, 51; dutasteride, 48) samples displayed the following Gleason patterns: 3+3 = 6 in 80 (54.8%) (placebo, 39; dutasteride, 41), 3+4 = 7 in 18 (12.3%) (placebo, 11; dutasteride, 7), and 4+4 = 8 in 1(0.68%) (placebo, 1). The remaining 47 samples showed atypical glands in 5 (3.4%) (placebo, 2; dutasteride, 3), HGPIN in 9 (6.1%) (placebo, 5; dutasteride, 4), and benign in 33 (22.6%) (placebo, 15; dutasteride, 18). Immunohistochemistry findings were positive for ERG and SPINK1 in 56 (56%) (placebo, 31; dutasteride, 25) and 9 (6.1%) (placebo, 5; dutasteride, 4) cases, respectively. ETV1 and ETV4 rearrangements were noted in 2 cases (1.4%) (placebo, 1; dutasteride, 1) and 1 (0.7%) (placebo, 1) case, respectively. No significant differences in the incidence of prostate cancer molecular aberrations between the groups were observed. There was no evidence that ETS fusion status was associated with disease progression.


Subject(s)
Gene Rearrangement , Oncogene Proteins, Fusion/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , 5-alpha Reductase Inhibitors/therapeutic use , Adenovirus E1A Proteins/genetics , Biopsy , Chromosomes, Artificial, Bacterial , Clinical Trials as Topic , DNA-Binding Proteins/genetics , Disease Progression , Dutasteride/therapeutic use , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ets , Transcription Factors/genetics
2.
Neoplasia ; 12(7): 590-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20651988

ABSTRACT

TMPRSS2-ERG gene fusions occur in 50% of prostate cancers and result in the overexpression of a chimeric fusion transcript that encodes a truncated ERG product. Previous attempts to detect truncated ERG products have been hindered by a lack of specific antibodies. Here, we characterize a rabbit anti-ERG monoclonal antibody (clone EPR 3864; Epitomics, Burlingame, CA) using immunoblot analysis on prostate cancer cell lines, synthetic TMPRSS2-ERG constructs, chromatin immunoprecipitation, and immunofluorescence. We correlated ERG protein expression with the presence of ERG gene rearrangements in prostate cancer tissues using a combined immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) analysis. We independently evaluated two patient cohorts and observed ERG expression confined to prostate cancer cells and high-grade prostatic intraepithelial neoplasia associated with ERG-positive cancer, as well as vessels and lymphocytes (where ERG has a known biologic role). Image analysis of 131 cases demonstrated nearly 100% sensitivity for detecting ERG rearrangement prostate cancer, with only 2 (1.5%) of 131 cases demonstrating strong ERG protein expression without any known ERG gene fusion. The combined pathology evaluation of 207 patient tumors for ERG protein expression had 95.7% sensitivity and 96.5% specificity for determining ERG rearrangement prostate cancer. In conclusion, this study qualifies a specific anti-ERG antibody and demonstrates exquisite association between ERG gene rearrangement and truncated ERG protein product expression. Given the ease of performing IHC versus FISH, ERG protein expression may be useful for molecularly subtyping prostate cancer based on ERG rearrangement status and suggests clinical utility in prostate needle biopsy evaluation.


Subject(s)
Antibodies, Monoclonal/pharmacology , DNA Mutational Analysis/methods , Oncogene Proteins, Fusion/immunology , Prostatic Intraepithelial Neoplasia/genetics , Prostatic Neoplasms/genetics , Trans-Activators/immunology , Adult , Aged , Animals , Biopsy, Needle , Case-Control Studies , Chromosome Aberrations , Cohort Studies , Fluorescent Antibody Technique , Humans , In Situ Hybridization, Fluorescence/methods , Male , Middle Aged , Oncogene Proteins, Fusion/analysis , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/classification , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Rabbits , Trans-Activators/analysis , Trans-Activators/genetics , Trans-Activators/metabolism , Transcriptional Regulator ERG
3.
Nat Med ; 16(7): 793-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20526349

ABSTRACT

Although recurrent gene fusions involving erythroblastosis virus E26 transformation-specific (ETS) family transcription factors are common in prostate cancer, their products are considered 'undruggable' by conventional approaches. Recently, rare targetable gene fusions involving the anaplastic lymphoma receptor tyrosine kinase (ALK) gene, have been identified in 1-5% of lung cancers, suggesting that similar rare gene fusions may occur in other common epithelial cancers, including prostate cancer. Here we used paired-end transcriptome sequencing to screen ETS rearrangement-negative prostate cancers for targetable gene fusions and identified the SLC45A3-BRAF (solute carrier family 45, member 3-v-raf murine sarcoma viral oncogene homolog B1) and ESRP1-RAF1 (epithelial splicing regulatory protein-1-v-raf-1 murine leukemia viral oncogene homolog-1) gene fusions. Expression of SLC45A3-BRAF or ESRP1-RAF1 in prostate cells induced a neoplastic phenotype that was sensitive to RAF and mitogen-activated protein kinase kinase (MAP2K1) inhibitors. Screening a large cohort of patients, we found that, although rare, recurrent rearrangements in the RAF pathway tend to occur in advanced prostate cancers, gastric cancers and melanoma. Taken together, our results emphasize the key role of RAF family gene rearrangements in cancer, suggest that RAF and MEK inhibitors may be useful in a subset of gene fusion-harboring solid tumors and demonstrate that sequencing of tumor transcriptomes and genomes may lead to the identification of rare targetable fusions across cancer types.


Subject(s)
Melanoma/genetics , Oncogene Proteins, Fusion/genetics , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-raf/genetics , RNA-Binding Proteins/genetics , Stomach Neoplasms/genetics , Translocation, Genetic , Humans , Male , Membrane Transport Proteins/genetics , Monosaccharide Transport Proteins , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Signal Transduction/genetics
4.
Am J Surg Pathol ; 34(4): 478-85, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20220513

ABSTRACT

BACKGROUND: Atypical cribriform lesions (ACLs) of the prostate consist of cribriform glands lined with cytologically malignant cells with partial or complete basal cell lining. It may represent cribriform "high-grade prostatic intraepithelial neoplasia" (HGPIN) or "intraductal carcinoma of the prostate" (IDC-P), which is almost always associated with clinically aggressive prostate carcinoma (PCa). Distinction between these 2 lesions has profound clinical significance, especially on needle biopsies. However, there are lesions that do not fully satisfy the criteria for IDC-P yet are worse than typical HGPIN and are difficult to distinguish based on morphologic criteria alone. METHODS: To better understand the biologic and molecular basis of distinction between cribriform HGPIN and IDC, we used break-apart fluorescence in-situ hybridization assay to assess ETS gene aberrations, a specific and commonest molecular alteration involving PCa, in a cohort of 16 isolated ACL, presumed to be an isolated cribriform HGPIN, and 45 carcinoma-associated ACL (ACL-PCa) on radical prostatectomy specimens, presumed to be spectrum of IDC-P. The latter was further divided into 2 groups: group A with marked nuclear atypia (nuclear size 6xnormal or larger) and/or comedonecrosis (n=21) and group B that did not fulfill these criteria (n=24). RESULTS: Overall, ERG rearrangement was absent (0 of 16) in isolated cribriform HGPIN, whereas present in 75% (36 of 48) of IDC-P, of which 65% (23 of 36) were through deletion and 35% (13 of 36) through insertion. Notably, 17% (6 of 36) of the IDC-P showed duplication of ERG rearrangement in combination with deletion of 5'-ERG. Hundred percent (34 of 34) of the IDC-P showed concordance of ERG rearrangement status with adjacent invasive carcinoma. There was no difference between the 2 groups of IDC-P lesions regarding prevalence of ERG rearrangement (group A 79% vs. group B 74%) and EDel2+ (20% vs. 15%). No case with ETV1, ETV4, or ETV5 rearrangement was identified. CONCLUSIONS: Our molecular data suggest that isolated cribriform HGPIN and IDC-P are biologically distinct lesions. Majority of ACL-PCa most likely represent intraductal spread of PCa. There is a significant overlap between IDC-P and HGPIN at the lower grade morphologic spectrum. ERG break-apart fluorescence in-situ hybridization assay provides insight into understanding the molecular basis of cribriform HGPIN and IDC-P and has potential clinical implications in their distinction on needle biopsies.


Subject(s)
Carcinoma, Ductal/pathology , Chromosome Aberrations , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-ets/genetics , Biomarkers, Tumor , Biopsy, Needle , Carcinoma, Ductal/genetics , Carcinoma, Ductal/surgery , Diagnosis, Differential , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization, Fluorescence , Male , Prostatic Intraepithelial Neoplasia/genetics , Prostatic Intraepithelial Neoplasia/surgery , Prostatic Neoplasms/genetics , Prostatic Neoplasms/surgery
5.
Mod Pathol ; 22(8): 1083-93, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19407851

ABSTRACT

The link between ERG rearrangement and PTEN (phosphatase and tensin homolog deleted on chromosome 10) deletion is unclear in prostate cancer progression. Using fluorescence in situ hybridization, we systematically validated the frequency and distribution of ERG and PTEN aberrations in a cohort of 73 benign prostate tissues, 59 high-grade prostatic intraepithelial neoplasia (HGPIN) foci, 281 localized prostate cancer and 47 androgen-independent metastatic prostate cancer patients. Overall, ERG rearrangement was present in 15% (5/33) of HGPIN, 45% (121/267) of localized cancers and 35% (15/43) of metastases. By contrast, PTEN deletion was identified in 9% (3/33) of HGPIN, 17% (42/251) of localized cancers and 54% (22/41) of metastases, of which 0%, 40% (17/42) and 45% (10/22) were homozygous, respectively. Concomitance of ERG rearrangement and PTEN deletion was observed in a subset of HGPIN. Significantly, association between PTEN deletion and ERG rearrangement was present both in localized cancers (P=0.0008) and metastases (P=0.02). Further, immunohistochemistry revealed significant correlation of decreased PTEN protein expression with PTEN genomic deletion both in localized and metastatic cancer. Of note, ERG aberration, but not PTEN deletion, was consistently identical both in localized cancer and adjacent HGPIN. Similarly, whereas all metastases (41/41, 100%) shared the same ERG status across multiple sites from the same patient, 5% (2/41) of cases showed discordance for PTEN deletion status across multiple sites. Collectively, our data support PTEN deletion as a late genetic event in human prostate cancer, presumably a 'second hit' after ERG rearrangement. PTEN deletion and ERG rearrangement may cooperate, but contribute at different stages, in prostate cancer progression.


Subject(s)
PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Trans-Activators/genetics , Disease Progression , Gene Deletion , Gene Rearrangement , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Tissue Array Analysis , Transcriptional Regulator ERG
6.
Mod Pathol ; 22(9): 1176-85, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19465903

ABSTRACT

Histologic variants of prostate carcinoma account for 5-10% of the disease and are typically seen in association with conventional acinar carcinoma. These variants often differ from the latter in clinical, immunophenotypic, and biologic potential. Recently, recurrent gene fusions between the androgen-regulated gene TMPRSS2 and the ETS transcription factors ERG, ETV1, ETV4, or ETV5 have been identified in a majority of conventional prostate carcinomas. However, the frequency and significance of this critical molecular event is unknown in the histologic variants of prostate carcinoma. Here, we used break-apart fluorescence in situ hybridization to assess TMPRSS2 and ETS aberrations in a series of select histologic variants: foamy gland carcinoma (N=17), ductal adenocarcinoma (N=18), mucinous carcinoma (N=18), and small cell carcinoma (N=7). A histologic variation of acinar adenocarcinoma, demonstrating glomeruloid morphology (N=9), was also investigated. Overall, 55% of histologic variant or variation morphologies demonstrated ETS aberrations (ERG in 54% and ETV1 in 1%). TMPRSS2:ERG fusion was identified in 83% (15/18), 71% (5/7), 50% (9/18), 33% (3/9), and 29% (5/17) of mucinous, small cell, ductal, glomeruloid, and foamy gland prostate carcinomas, respectively. Previously, we reported that 100% of androgen-independent metastatic prostate carcinomas harboring TMPRSS2:ERG gene fusion were associated with interstitial deletion (Edel). Interestingly, ERG rearrangement in small cell carcinomas occurred exclusively through Edel, supporting the notion that TMPRSS2:ERG with Edel is an aggressive molecular subtype. SPINK1, a biomarker expressed exclusively in a subset of ETS negative prostate carcinomas, was expressed in 6% of ETS negative histologic variants, specifically in ductal adenocarcinoma. Notably, 88% (43/49) variant morphologies in this cohort showed concordance of TMPRSS2:ERG fusion with associated conventional acinar type, suggesting that variant morphology is clonally related to the latter. Overall, our data provide insight into the origin, molecular mechanism, and phenotypic association of ETS fusions in histologic variants of prostate carcinoma.


Subject(s)
Carcinoma/genetics , Carcinoma/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-ets/genetics , Aged , Biomarkers, Tumor/analysis , Carrier Proteins/biosynthesis , Chromosome Aberrations , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Neoplasm Staging , Oncogene Proteins, Fusion/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis , Trypsin Inhibitor, Kazal Pancreatic
SELECTION OF CITATIONS
SEARCH DETAIL
...