Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 17 de 17
Filtrer
1.
Oncotarget ; 7(4): 3897-904, 2016 Jan 26.
Article de Anglais | MEDLINE | ID: mdl-26684029

RÉSUMÉ

Prostate cancer is notorious for its heterogeneity, which poses a problem for the applicability of diagnostic molecular markers. However, heterogeneity analysis can provide valuable information on the chronology in which molecular alterations arise. Here, we constructed a heterogeneity tissue microarray (TMA) comprising samples from 10 different tumor areas of 189 prostate cancers each in order to study the sequence of two frequent molecular alterations, i.e. 6q15 deletion and TMPRSS2:ERG fusion. Previous work shows a marked inverse relationship between these alterations, suggesting that presence of one of these alterations might impact development of the other. 6q15 deletion was analyzed by fluorescence in situ hybridization and ERG-expression by immunohistochemistry. Only 6.6% of 334 ERG-positive but 28.4% of 440 ERG-negative TMA spots showed 6q15 deletions (p < 0.0001). A breakdown of these data to the level of tumor foci revealed 6q deletions in 138 tumor foci that were large enough to have at least 3 analyzable TMA spots. These included 42 tumor foci with homogeneous ERG positivity and 16 with homogeneous 6q15 deletions. Remarkably, six of the 42 homogeneously ERG-positive tumor foci (14.3%) harbored small 6q15-deleted areas, but none of the 34 6q15-deleted foci showed areas of ERG positivity (p = 0.022). In conclusion, our data suggest that ERG-fusion can precede 6q15 deletion, but not vice versa. The complete absence of ERG-positive tumor areas in 6q15-deleted tumor foci further suggest that the functional consequences of 6q15 deletions may prevent the development of TMPRSS2:ERG fusions.


Sujet(s)
Adénocarcinome/génétique , Chromosomes humains de la paire 6/génétique , Délétion de gène , Protéines de fusion oncogènes/génétique , Tumeurs de la prostate/génétique , Transactivateurs/génétique , Adénocarcinome/métabolisme , Adénocarcinome/secondaire , Sujet âgé , Sujet âgé de 80 ans ou plus , Humains , Techniques immunoenzymatiques , Hybridation fluorescente in situ , Métastase lymphatique , MAP Kinase Kinase Kinases/métabolisme , Mâle , Adulte d'âge moyen , Grading des tumeurs , Stadification tumorale , Pronostic , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Analyse sur puce à tissus , Régulateur transcriptionnel ERG
2.
Int J Oncol ; 46(4): 1637-42, 2015 Apr.
Article de Anglais | MEDLINE | ID: mdl-25625310

RÉSUMÉ

Prostate cancer is characterized by structural rearrangements, most frequently including translocations between androgen-dependent genes and members of the ETS family of transcription factor like TMPRSS2:ERG. In a recent whole genome sequencing study we identified 140 gene fusions that were unrelated to ETS genes in 11 prostate cancers. The aim of the present study was to estimate the prevalence of non-ETS gene fusions. We randomly selected 27 of these rearrangements and analyzed them by fluorescence in situ hybridization (FISH) in a tissue microarray format containing 500 prostate cancers. Using break-apart FISH probes for one fusion partner each, we found rearrangements of 13 (48%) of the 27 analyzed genes in 300-400 analyzable cancers per gene. Recurrent breakage, often accompanied by partial deletion of the genes, was found for NCKAP5, SH3BGR and TTC3 in 3 (0.8%) tumors each, as well as for ARNTL2 and ENOX1 in 2 (0.5%) cancers each. One rearranged tumor sample was observed for each of VCL, ZNF578, IMMP2L, SLC16A12, PANK1, GPHN, LRP1 and ZHX2. Balanced rearrangements, indicating possible gene fusion, were found for ZNF578, SH3BGR, LPR12 and ZHX2 in individual cancers only. The results of the present study confirm that rearrangements involving non-ETS genes occur in prostate cancer, but demonstrate that they are highly individual and typically non-recurrent.


Sujet(s)
Aberrations des chromosomes , Chromosomes humains/génétique , Protéines de fusion oncogènes/génétique , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Adulte , Sujet âgé , Humains , Hybridation fluorescente in situ/méthodes , Mâle , Adulte d'âge moyen , Protéines proto-oncogènes c-ets/génétique , Analyse sur puce à tissus/méthodes
3.
Eur Urol ; 66(6): 978-81, 2014 Dec.
Article de Anglais | MEDLINE | ID: mdl-25015038

RÉSUMÉ

Based on next-generation sequencing of early-onset prostate cancer (PCa), we earlier demonstrated that PCa in young patients is prone to rearrangements involving androgen-regulated genes-such as transmembrane protease, serine 2 (TMPRSS2)-v-ets avian erythroblastosis virus E26 oncogene homolog (ERG) fusion-and provided data suggesting that this situation might be caused by increased androgen signaling in younger men. In the same study, an accumulation of chromosomal deletions was found in cancers of elderly patients. To determine how age-dependent molecular features relate to cancer phenotype, an existing data set of 11,152 PCas was expanded by additional fluorescence in situ hybridization analyses of phosphatase and tensin homolog (PTEN), 6q15 and 5q21. The results demonstrate that the decrease in TMPRSS2-ERG fusions with increasing patient age is limited to low-grade cancers (Gleason ≤3+4) and that the significant increase in the deletion frequency with age was strictly limited to ERG-negative cancers for 6q15 and 5q21 but to ERG-positive cancers for PTEN. These data suggest that the accumulation of non-androgen-linked genomic alterations with advanced patient age may require an appropriate microenvironment, such as a positive or negative ERG status. The strong link of ERG activation to young patient age and low-grade cancers may help to explain a slight predominance of low-grade cancers in young patients.


Sujet(s)
Fusion de gènes , Protéines de fusion oncogènes/génétique , Phosphohydrolase PTEN/génétique , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Transactivateurs/génétique , Adulte , Facteurs âges , Sujet âgé , Sujet âgé de 80 ans ou plus , Séquence nucléotidique , Chromosomes humains de la paire 5 , Chromosomes humains de la paire 6 , Instabilité du génome , Humains , Hybridation fluorescente in situ , Mâle , Adulte d'âge moyen , Grading des tumeurs , Tumeurs de la prostate/sang , Délétion de séquence , Testostérone/sang , Régulateur transcriptionnel ERG
4.
Mod Pathol ; 27(12): 1612-20, 2014 Dec.
Article de Anglais | MEDLINE | ID: mdl-24762546

RÉSUMÉ

TMPRSS2:ERG fusions, in combination with deletion of the phosphatase and tensin homolog (PTEN) tumor suppressor, have been suggested to cooperatively drive tumor progression in prostate cancer. We utilized a novel heterogeneity tissue microarray containing samples from 10 different tumor blocks of 189 prostatectomy specimens to study heterogeneity of genomic PTEN alterations in individual foci. PTEN alterations were found in 48/123 (39%) analyzable individual tumor foci, including 40 foci with deletions, 7 with deletion and rearrangement, and 1 focus with rearrangement only. PTEN was homogeneously aberrant in only 4 (8%) and heterogeneously in 44 (92%) of the foci. We found a specific sequence of molecular events from PTEN breakage followed by deletion of DNA sequences flanking the breakpoint, resulting in homozygous deletion. The observation that 16 of 19 foci with homogeneous ERG positivity had focal PTEN alterations but none of 10 foci with PTEN alterations had focal ERG positivity (P<0.0001) suggests that PTEN alterations typically develop subsequent to ERG fusions. We demonstrate a high level of intratumoral heterogeneity of PTEN alterations with deletions and rearrangements that challenges potential PTEN routine diagnosis testing in biopsies. The observation that PTEN alterations develop subsequent to ERG fusion strongly suggests that ERG expression may directly drive development of PTEN aberrations.


Sujet(s)
Adénocarcinome/génétique , Protéines de fusion oncogènes/génétique , Phosphohydrolase PTEN/génétique , Tumeurs de la prostate/génétique , Transactivateurs/génétique , Délétion de gène , Humains , Immunohistochimie , Hybridation fluorescente in situ , Mâle , Analyse sur puce à tissus , Régulateur transcriptionnel ERG
5.
Int J Cancer ; 135(6): 1369-80, 2014 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-24523142

RÉSUMÉ

Despite a multitude of p53 immunohistochemistry (IHC) studies, data on the combined effect of nuclear p53 protein accumulation and TP53 genomic inactivation are lacking for prostate cancer. A tissue microarray including 11,152 prostate cancer samples was analyzed by p53 IHC and fluorescence in situ hybridization. Nuclear p53 accumulation was found in 10.1% of patients including 1.4% with high-level and 8.7% with low-level immunostaining. TP53 sequencing revealed that 17 of 22 (77%) cases with high-level p53 immunostaining, but only 3% (1 of 31) low-level p53 cases carried putative dominant-negative mutations. TP53 deletions occurred in 14.8% of cancers. Both deletions and protein accumulation were linked to unfavorable tumor phenotype and prostate specific antigen (PSA) recurrence (p<0.0001 each). The combination of both methods revealed subgroups with remarkable differences in their clinical course. Tumors with either TP53 deletion (14%) or low-level p53 positivity (8.7%) had identical risks of PSA recurrence, which were markedly higher than in cancers without p53 alterations (p<0.0001). Tumors with both p53 deletion and low-level p53 positivity (1.5%) had a worse prognosis than patients with only one of these alterations (p<0.0001). Tumors with strong p53 immunostaining or homozygous inactivation through deletion of one allele and disrupting translocation involving the second allele had the worst outcome, independent from clinical and pathological parameters. These data demonstrate a differential clinical impact of various TP53 alterations in prostate cancer. Strong p53 immunostaining-most likely accompanying dominant negative or oncogenic p53 mutation-has independent prognostic relevance and may thus represent a clinical useful molecular feature of prostate cancer.


Sujet(s)
Gènes p53 , Tumeurs de la prostate/génétique , Aberrations des chromosomes , Extinction de l'expression des gènes , Humains , Immunohistochimie , Hybridation fluorescente in situ , Kallicréines/métabolisme , Mâle , Analyse multifactorielle , Modèles des risques proportionnels , Antigène spécifique de la prostate/métabolisme , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/chirurgie , Analyse sur puce à tissus
6.
Clin Oral Investig ; 18(1): 211-7, 2014 Jan.
Article de Anglais | MEDLINE | ID: mdl-23404560

RÉSUMÉ

OBJECTIVES: The present study aims to investigate the relevance of immunohistochemical p53 expression in carcinomas of the oral cavity and of the head and neck region. Long-term clinical and histopathological follow-up findings as well as HPV status are correlated with the results of this examination. MATERIALS AND METHODS: Sections made from two tissue arrays composed of 222 oral squamous cell carcinomas and 427 squamous cell carcinomas of the head and neck region, respectively, were examined for p53 expression and Ki-67 index by means of immunohistochemistry. Correlation of long-term clinical findings of the patients and pathological features of tumours with laboratory results were examined statistically. RESULTS: No significant correlation was found between the p53 immunohistochemical expression in the 207 oral carcinomas and features of the tumours and patient outcomes. There was no significant association between the Ki-67 labelling index and the p53 expression. DISCUSSION: Our failure in detecting any association of the p53 immunohistochemical expression regardless of HPV status with clinical features of these tumours suggests it lacks a prognostic value for squamous cell carcinomas of the oral cavity. CLINICAL RELEVANCE: The prognostic value of p53 immunostaining in oral squamous carcinoma is not clarified yet. In the present study, there is no impact on any prognostical item nor even a correlation with cell proliferation (Ki-67) regardless of HPV status.


Sujet(s)
Carcinome épidermoïde/métabolisme , Tumeurs de la tête et du cou/métabolisme , Tumeurs de la bouche/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Alphapapillomavirus/isolement et purification , Séquence nucléotidique , Carcinome épidermoïde/anatomopathologie , Amorces ADN , Femelle , Tumeurs de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/virologie , Humains , Immunohistochimie , Mâle , Tumeurs de la bouche/anatomopathologie , Tumeurs de la bouche/virologie , Réaction de polymérisation en chaîne , Pronostic
7.
J Pathol ; 231(1): 130-41, 2013 Sep.
Article de Anglais | MEDLINE | ID: mdl-23794398

RÉSUMÉ

Deletion of 3p13 has been reported from about 20% of prostate cancers. The clinical significance of this alteration and the tumour suppressor gene(s) driving the deletion remain to be identified. We have mapped the 3p13 deletion locus using SNP array analysis and performed fluorescence in situ hybridization (FISH) analysis to search for associations between 3p13 deletion, prostate cancer phenotype and patient prognosis in a tissue microarray containing more than 3200 prostate cancers. SNP array analysis of 72 prostate cancers revealed a small deletion at 3p13 in 14 (19%) of the tumours, including the putative tumour suppressors FOXP1, RYBP and SHQ1. FISH analysis using FOXP1-specific probes revealed deletions in 16.5% and translocations in 1.2% of 1828 interpretable cancers. 3p13 deletions were linked to adverse features of prostate cancer, including advanced stage (p < 0.0001), high Gleason grade (p = 0.0125), and early PSA recurrence (p = 0.0015). In addition, 3p13 deletions were linked to ERG(+) cancers and to PTEN deletions (p < 0.0001 each). A subset analysis of ERG(+) tumours revealed that 3p13 deletions occurred independently from PTEN deletions (p = 0.3126), identifying tumours with 3p13 deletion as a distinct molecular subset of ERG(+) cancers. mRNA expression analysis confirmed that all 3p13 genes were down regulated by the deletion. Ectopic over-expression of FOXP1, RYBP and SHQ1 resulted in decreased colony-formation capabilities, corroborating a tumour suppressor function for all three genes. In summary, our data show that deletion of 3p13 defines a distinct and aggressive molecular subset of ERG(+) prostate cancers, which is possibly driven by inactivation of multiple tumour suppressors.


Sujet(s)
Adénocarcinome/génétique , Délétion de segment de chromosome , Chromosomes humains de la paire 3/génétique , Gènes suppresseurs de tumeur , Tumeurs de la prostate/génétique , Adénocarcinome/métabolisme , Adénocarcinome/mortalité , Adénocarcinome/anatomopathologie , Lignée cellulaire tumorale , Facteurs de transcription Forkhead/génétique , Facteurs de transcription Forkhead/métabolisme , Analyse de profil d'expression de gènes , Techniques de knock-down de gènes , Allemagne/épidémiologie , Humains , Estimation de Kaplan-Meier , Mâle , Récidive tumorale locale , Séquençage par oligonucléotides en batterie , Protéines de fusion oncogènes/métabolisme , Polymorphisme de nucléotide simple , Prostate/métabolisme , Prostate/anatomopathologie , Prostatectomie , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/mortalité , Tumeurs de la prostate/anatomopathologie , Protéines de répression/génétique , Protéines de répression/métabolisme , Analyse sur puce à tissus
8.
Cancer Res ; 73(9): 2795-805, 2013 May 01.
Article de Anglais | MEDLINE | ID: mdl-23492366

RÉSUMÉ

Deletions involving the chromosomal band 5q21 are among the most frequent alterations in prostate cancer. Using single-nucleotide polymorphism (SNP) arrays, we mapped a 1.3 megabase minimally deleted region including only the repulsive guidance molecule B (RGMB) and chromodomain helicase DNA-binding protein 1 (CHD1) genes. Functional analyses showed that CHD1 is an essential tumor suppressor. FISH analysis of 2,093 prostate cancers revealed a strong association between CHD1 deletion, prostate-specific antigen (PSA) biochemical failure (P = 0.0038), and absence of ERG fusion (P < 0.0001). We found that inactivation of CHD1 in vitro prevents formation of ERG rearrangements due to impairment of androgen receptor (AR)-dependent transcription, a prerequisite for ERG translocation. CHD1 is required for efficient recruitment of AR to responsive promoters and regulates expression of known AR-responsive tumor suppressor genes, including NKX3-1, FOXO1, and PPARγ. Our study establishes CHD1 as the 5q21 tumor suppressor gene in prostate cancer and shows a key role of this chromatin remodeling factor in prostate cancer biology.


Sujet(s)
Helicase/génétique , Protéines de liaison à l'ADN/génétique , Réarrangement des gènes , Gènes suppresseurs de tumeur , Tumeurs de la prostate/génétique , Transactivateurs/métabolisme , Lignée cellulaire tumorale , Chromosomes humains de la paire 5 , Délétion de gène , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux , Humains , Hybridation fluorescente in situ , Mâle , Séquençage par oligonucléotides en batterie , Polymorphisme de nucléotide simple , Antigène spécifique de la prostate/métabolisme , Récepteurs aux androgènes/métabolisme , Régulateur transcriptionnel ERG , Translocation génétique
9.
Mod Pathol ; 26(7): 975-83, 2013 Jul.
Article de Anglais | MEDLINE | ID: mdl-23370768

RÉSUMÉ

6q12-22 is the second most commonly deleted genomic region in prostate cancer. Mapping studies have described a minimally deleted area at 6q15, containing MAP3K7/TAK1, which was recently shown to have tumor suppressive properties. To determine prevalence and clinical significance of MAP3K7 alterations in prostate cancer, a tissue microarray containing 4699 prostate cancer samples was analyzed by fluorescence in situ hybridization. Heterozygous MAP3K7 deletions were found in 18.48% of 2289 interpretable prostate cancers. MAP3K7 deletions were significantly associated with advanced tumor stage (P<0.0001), high Gleason grade (P<0.0001), lymph node metastasis (P<0.0108) and early biochemical recurrence (P<0.0001). MAP3K7 alterations were typically limited to the loss of one allele as homozygous deletions were virtually absent and sequencing analyses revealed no evidence for MAP3K7 mutations in 15 deleted and in 14 non-deleted cancers. There was a striking inverse association of MAP3K7 deletions and TMPRSS2:ERG fusion status with 26.7% 6q deletions in 1125 ERG-negative and 11.1% 6q deletions in 1198 ERG-positive cancers (P<0.0001). However, the strong prognostic role of 6q deletions was retained in both ERG-positive and ERG-negative cancers (P<0.0001 each). In summary, our study identifies MAP3K7 deletion as a prominent feature in ERG-negative prostate cancer with strong association to tumor aggressiveness. MAP3K7 alterations are typically limited to one allele of the gene. Together with the demonstrated tumor suppressive function in cell line experiments and lacking evidence for inactivation through hypermethylation, these results indicate MAP3K7 as a gene for which haploinsufficency is substantially tumorigenic.


Sujet(s)
MAP Kinase Kinase Kinases/génétique , Récidive tumorale locale/génétique , Tumeurs de la prostate/génétique , Adulte , Sujet âgé , Marqueurs biologiques tumoraux/génétique , Technique de Western , Chromosomes humains de la paire 6 , Délétion de gène , Humains , Hybridation fluorescente in situ , MAP Kinase Kinase Kinases/sang , Mâle , Adulte d'âge moyen , Récidive tumorale locale/anatomopathologie , Protéines de fusion oncogènes/génétique , Réaction de polymérisation en chaîne , Antigène spécifique de la prostate/sang , Tumeurs de la prostate/anatomopathologie , Analyse sur puce à tissus
10.
PLoS One ; 8(2): e54873, 2013.
Article de Anglais | MEDLINE | ID: mdl-23393560

RÉSUMÉ

Chromosome 8q24 is the most commonly amplified region across multiple cancer types, and the typical length of the amplification suggests that it may target additional genes to MYC. To explore the roles of the genes most frequently included in 8q24 amplifications, we analyzed the relation between copy number alterations and gene expression in three sets of endometrial cancers (N = 252); and in glioblastoma, ovarian, and breast cancers profiled by TCGA. Among the genes neighbouring MYC, expression of the bromodomain-containing gene ATAD2 was the most associated with amplification. Bromodomain-containing genes have been implicated as mediators of MYC transcriptional function, and indeed ATAD2 expression was more closely associated with expression of genes known to be upregulated by MYC than was MYC itself. Amplifications of 8q24, expression of genes downstream from MYC, and overexpression of ATAD2 predicted poor outcome and increased from primary to metastatic lesions. Knockdown of ATAD2 and MYC in seven endometrial and 21 breast cancer cell lines demonstrated that cell lines that were dependent on MYC also depended upon ATAD2. These same cell lines were also the most sensitive to the histone deacetylase (HDAC) inhibitor Trichostatin-A, consistent with prior studies identifying bromodomain-containing proteins as targets of inhibition by HDAC inhibitors. Our data indicate high ATAD2 expression is a marker of aggressive endometrial cancers, and suggest specific inhibitors of ATAD2 may have therapeutic utility in these and other MYC-dependent cancers.


Sujet(s)
Adenosine triphosphatases/métabolisme , Protéines de liaison à l'ADN/métabolisme , Tumeurs de l'endomètre/génétique , Tumeurs de l'endomètre/métabolisme , Gènes myc/physiologie , Génomique/méthodes , ATPases associated with diverse cellular activities , Adenosine triphosphatases/génétique , Lignée cellulaire tumorale , Protéines de liaison à l'ADN/génétique , Femelle , Gènes myc/génétique , Humains , Immunotransfert , Hybridation fluorescente in situ
11.
Cancer Cell ; 23(2): 159-70, 2013 Feb 11.
Article de Anglais | MEDLINE | ID: mdl-23410972

RÉSUMÉ

Early-onset prostate cancer (EO-PCA) represents the earliest clinical manifestation of prostate cancer. To compare the genomic alteration landscapes of EO-PCA with "classical" (elderly-onset) PCA, we performed deep sequencing-based genomics analyses in 11 tumors diagnosed at young age, and pursued comparative assessments with seven elderly-onset PCA genomes. Remarkable age-related differences in structural rearrangement (SR) formation became evident, suggesting distinct disease pathomechanisms. Whereas EO-PCAs harbored a prevalence of balanced SRs, with a specific abundance of androgen-regulated ETS gene fusions including TMPRSS2:ERG, elderly-onset PCAs displayed primarily non-androgen-associated SRs. Data from a validation cohort of > 10,000 patients showed age-dependent androgen receptor levels and a prevalence of SRs affecting androgen-regulated genes, further substantiating the activity of a characteristic "androgen-type" pathomechanism in EO-PCA.


Sujet(s)
Réarrangement des gènes , Génomique , Protéines de fusion oncogènes/génétique , Tumeurs de la prostate/génétique , Récepteurs aux androgènes/génétique , Serine endopeptidases/génétique , Transactivateurs/génétique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Biologie informatique , Séquençage nucléotidique à haut débit , Humains , Mâle , Adulte d'âge moyen , Régulateur transcriptionnel ERG
12.
Am J Pathol ; 181(2): 401-12, 2012 Aug.
Article de Anglais | MEDLINE | ID: mdl-22705054

RÉSUMÉ

The phosphatase and tensin homolog deleted on chromosome 10 (PTEN) gene is often altered in prostate cancer. To determine the prevalence and clinical significance of the different mechanisms of PTEN inactivation, we analyzed PTEN deletions in TMAs containing 4699 hormone-naïve and 57 hormone-refractory prostate cancers using fluorescence in situ hybridization analysis. PTEN mutations and methylation were analyzed in subsets of 149 and 34 tumors, respectively. PTEN deletions were present in 20.2% (458/2266) of prostate cancers, including 8.1% heterozygous and 12.1% homozygous deletions, and were linked to advanced tumor stage (P < 0.0001), high Gleason grade (P < 0.0001), presence of lymph node metastasis (P = 0.0002), hormone-refractory disease (P < 0.0001), presence of ERG gene fusion (P < 0.0001), and nuclear p53 accumulation (P < 0.0001). PTEN deletions were also associated with early prostate-specific antigen recurrence in univariate (P < 0.0001) and multivariate (P = 0.0158) analyses. The prognostic impact of PTEN deletion was seen in both ERG fusion-positive and ERG fusion-negative tumors. PTEN mutations were found in 4 (12.9%) of 31 cancers with heterozygous PTEN deletions but in only 1 (2%) of 59 cancers without PTEN deletion (P = 0.027). Aberrant PTEN promoter methylation was not detected in 34 tumors. The results of this study demonstrate that biallelic PTEN inactivation, by either homozygous deletion or deletion of one allele and mutation of the other, occurs in most PTEN-defective cancers and characterizes a particularly aggressive subset of metastatic and hormone-refractory prostate cancers.


Sujet(s)
Délétion de gène , Protéines de fusion oncogènes/métabolisme , Phosphohydrolase PTEN/génétique , Antigène spécifique de la prostate/métabolisme , Tumeurs de la prostate/enzymologie , Tumeurs de la prostate/anatomopathologie , Transactivateurs/métabolisme , Sujet âgé , Marqueurs biologiques tumoraux/métabolisme , Chromosomes humains de la paire 10/génétique , Méthylation de l'ADN/génétique , Analyse de mutations d'ADN , Évolution de la maladie , Épigenèse génétique , Génome humain/génétique , Humains , Immunohistochimie , Mâle , Adulte d'âge moyen , Analyse multifactorielle , Phosphohydrolase PTEN/métabolisme , Phénotype , Polymorphisme de nucléotide simple/génétique , Régions promotrices (génétique)/génétique , Modèles des risques proportionnels , Récidive , Régulateur transcriptionnel ERG , Protéine p53 suppresseur de tumeur/métabolisme
13.
J Clin Pathol ; 65(8): 693-8, 2012 Aug.
Article de Anglais | MEDLINE | ID: mdl-22639407

RÉSUMÉ

BACKGROUND AND AIM: Gastric carcinoma is the second most frequent cause of cancer-related death worldwide. As PTEN is a potential modifier of tumour response to trastuzumab, a recently approved therapy in metastatic HER2 positive gastric cancer, the existence of PTEN deletions in primary gastric cancer was investigated. METHODS: 230 primary gastric cancers were analysed in a tissue microarray format by dual labelling fluorescence in situ hybridisation for PTEN deletion. HER2 analysis was also performed. To study PTEN deletion heterogeneity, all available large tissue sections from primary cancer and corresponding metastases were analysed in seven patients. RESULTS: Eight of 180 interpretable primary gastric cancer spots showed PTEN deletions (4.4%), including seven hemizygous and one homozygous deletion. PTEN deletion was correlated with nodal (8 of 122 cases (6.6%); p=0.041) and distant metastases (4 of 19 (21.1%); p<0.001). Large section validation showed a homogeneous distribution of PTEN deletion. HER2 positivity was seen in one PTEN deleted case. CONCLUSION: Genomic PTEN deletion is a rare event in gastric adenocarcinoma but correlates with metastatic disease. The homogeneous distribution pattern indicates that this alteration occurs early in tumour development.


Sujet(s)
Adénocarcinome/génétique , Marqueurs biologiques tumoraux/génétique , Délétion de gène , Phosphohydrolase PTEN/génétique , Tumeurs de l'estomac/génétique , Adénocarcinome/enzymologie , Adénocarcinome/mortalité , Adénocarcinome/secondaire , Sujet âgé , Loi du khi-deux , Femelle , Prédisposition génétique à une maladie , Allemagne , Hémizygote , Homozygote , Humains , Immunohistochimie , Hybridation fluorescente in situ , Estimation de Kaplan-Meier , Mâle , Adulte d'âge moyen , Valeur prédictive des tests , Pronostic , Récepteur ErbB-2/analyse , Appréciation des risques , Facteurs de risque , Tumeurs de l'estomac/enzymologie , Tumeurs de l'estomac/mortalité , Tumeurs de l'estomac/anatomopathologie , Facteurs temps , Analyse sur puce à tissus/méthodes
14.
Prostate ; 72(8): 898-903, 2012 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-21956681

RÉSUMÉ

BACKGROUND: Loss of the Y chromosome is a frequently reported chromosomal abnormality in many tumor types. This study was undertaken to investigate the frequency of Y chromosome losses and this chromosomal abnormality might play a potential role in prostate cancer. METHODS: A preexisting prostate cancer tissue microarray (TMA) containing samples of 3,261 patients treated by radical prostatectomy with clinical follow-up data was used in this study. TMA sections were analyzed by fluorescence in situ hybridization (FISH) using a dual labeling probe for the centromeres of the X and Y chromosome. RESULTS: Unequivocal losses of the Y chromosome were seen in only 12 of 2,053 analyzable cases. No significant associations were found between Y loss and patient age, pT stage, and the risk of PSA recurrence. Interestingly, in our study the presence of Y losses was significantly associated with high Gleason grade (P = 0.0034). CONCLUSIONS: Loss of the Y chromosome is a rare event in prostate cancer. Y losses occur in much higher rates in most other cancer types. For this reason, we suggest that the expression of at least one Y chromosome gene is essential for prostate epithelial cells and it is possible that such a gene could represent a suitable target for future therapy of prostate cancer.


Sujet(s)
Vieillissement/génétique , Délétion de segment de chromosome , Chromosomes Y humains , Tumeurs de la prostate/génétique , Sujet âgé , Vieillissement/anatomopathologie , Centromère/anatomopathologie , Études de suivi , Humains , Hybridation fluorescente in situ , Mâle , Adulte d'âge moyen , Stadification tumorale , Prostatectomie , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/chirurgie , Études rétrospectives
15.
Clin Cancer Res ; 17(18): 5878-88, 2011 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-21791629

RÉSUMÉ

PURPOSE: About 50% of prostate cancers have TMPRSS2-ERG fusions with concurrent ERG overexpression. The aim of this study was to determine whether clinical differences exist between ERG-positive and ERG-negative cancers in surgically treated patients not exposed to antihormonal therapy. A secondary aim was to search for differences between these tumor classes. EXPERIMENTAL DESIGN: A tissue microarray containing samples from more than 2,800 prostate cancers with clinical data was analyzed for ERG alterations by immunohistochemistry and FISH. Results were compared with tumor phenotype, biochemical recurrence, and molecular features considered important for prostate cancer. The effect of ERG on androgen receptor (AR)-dependent transcription was analyzed in cell lines. RESULTS: ERG expression was found in 52.4% of 2,805 cancers with a 95% concordance between ERG expression and ERG gene rearrangement detected by FISH. ERG expression was unrelated to clinical outcome and tumor phenotype. Differences in AMACR, Annexin A3, Bcl2, CD10, ALCAM, chromogranin A, epidermal growth factor receptor, HER2, mTOR, p53, and synaptophysin status were significant but minimal in absolute numbers. The most striking difference was found for AR expression, which was markedly higher in ERG-positive cancers. In vitro studies showed ERG-dependent impairment of AR-mediated transcriptional activity. CONCLUSIONS: The striking similarities between these two types of prostate cancers rules out a major impact of ERG on tumor aggressiveness in early, not hormonally treated cancer. The marked difference in AR levels between ERG-positive and -negative cancers supports a systematic difference in potential response to hormonal therapy as previously observed in clinical trials.


Sujet(s)
Récidive tumorale locale/diagnostic , Antigène spécifique de la prostate/sang , Tumeurs de la prostate/diagnostic , Transactivateurs/génétique , Transactivateurs/métabolisme , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Lignée cellulaire tumorale , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux , Réarrangement des gènes , Humains , Mâle , Adulte d'âge moyen , Prostatectomie , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/chirurgie , Récepteurs aux androgènes/métabolisme , Régulateur transcriptionnel ERG
16.
BMC Med Genet ; 11: 106, 2010 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-20598160

RÉSUMÉ

BACKGROUND: Mutation analysis of KIT and PDGFRA genes in gastrointestinal stromal tumors is gaining increasing importance for prognosis of GISTs and for prediction of treatment response. Several groups have identified specific mutational subtypes in KIT exon 11 associated with an increased risk of metastatic disease whereas GISTs with PDGFRA mutations often behave less aggressive. Furthermore, in advanced GIST disease with proven KIT exon 9 mutation the doubled daily dose of 800 mg imatinib increases the progression free survival and is now recommended both in the European and the American Guidelines. In Germany, there are still no general rules how to perform mutational analysis. METHODS: When comparing results from six different molecular laboratories we recognized the need of standardisation. Six German university laboratories with experience in mutation analysis in GISTs joined together to develop recommendations for the mutation analysis of the most common and clinically relevant hot spots, i. e. KIT exons 9 and 11 and PDGFRA exon 18. We performed a three-phased interlaboratory trial to identify pitfalls in performing molecular analysis in GISTs. RESULTS: We developed a design for a continuous external laboratory trial. In 2009 this external trial was conducted by 19 laboratories via the initiative for quality assurance in pathology (QuiP) of the German Society of Pathology and the Professional Association of German Pathologists. CONCLUSIONS: By performing a three-phased internal interlaboratory trial and conducting an external trial in Germany we were able to identify potential pitfalls when performing KIT and PDGFRA mutational analysis in gastrointestinal stromal tumors. We developed standard operation procedures which are provided with the manuscript to allow other laboratories to prevent these pitfalls.


Sujet(s)
Tumeurs stromales gastro-intestinales/génétique , Tumeurs stromales gastro-intestinales/anatomopathologie , Mutation , Récepteur au PDGF alpha/génétique , Benzamides , Analyse de mutations d'ADN , Survie sans rechute , Exons , Tumeurs stromales gastro-intestinales/traitement médicamenteux , Allemagne , Humains , Mésilate d'imatinib , Pipérazines , Pronostic , Pyrimidines
17.
Genes Chromosomes Cancer ; 49(1): 1-8, 2010 Jan.
Article de Anglais | MEDLINE | ID: mdl-19787783

RÉSUMÉ

Recently, amplification of PPFIA1, encoding a member of the liprin family located about 600 kb telomeric to CCND1 on chromosome band 11q13, was described in squamous cell carcinoma of head and neck. Because 11q13 amplification is frequent in breast cancer, and PPFIA1 has been suggested to contribute to mammary gland development, we hypothesized that PPFIA1 might also be involved in the 11q13 amplicon in breast cancer and contribute to breast cancer development. A tissue microarray containing more than 2000 human breast cancers was analyzed for gene copy numbers of PPFIA1 and CCND1 by means of fluorescence in situ hybridization. PPFIA1 amplification was found in 248/1583 (15.4%) of breast cancers. Coamplification with CCND1 was found in all (248/248, 100%) PPFIA1-amplified cancers. CCND1 amplification without PPFIA1 coamplification was found in additional 117 (4.7%) tumors. Amplification of both PPFIA1 and CCND1 were significantly associated with high-grade phenotype (P = 0.0002) but were unrelated to tumor stage (P = 0.7066) or nodal stage (P = 0.5807). No difference in patient prognosis was found between 248 CCND1/PPFIA1 coamplified tumors and 117 tumors with CCND1 amplification alone (P = 0.6419). These data show that PPFIA1 amplification occurs frequently in breast cancer. The higher incidence of CCND1 amplification when compared with PPFIA1, the lack of prognostic relevance of coamplifications, and the fact that PPFIA1 amplification was found exclusively in CCND1-amplified cancers suggest that PPFIA1 gene copy number changes represent concurrent events of CCND1 amplification rather than specific biological incidents.


Sujet(s)
Protéines adaptatrices de la transduction du signal/génétique , Tumeurs du sein/génétique , Cycline D1/génétique , Amplification de gène , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Tumeurs du sein/anatomopathologie , Chromosomes humains de la paire 11 , Femelle , Dosage génique , Humains , Incidence , Adulte d'âge moyen , Phénotype , Pronostic , Analyse sur puce à tissus
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...