Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Lab Invest ; 101(1): 26-37, 2021 01.
Article in English | MEDLINE | ID: mdl-32873880

ABSTRACT

Most NUTM1-rearranged neoplasms (NRNs) have fusions between NUTM1 and BRD (bromodomain-containing) family members and are termed NUT carcinomas (NCs) because they show some squamous differentiation. However, some NRNs are associated with fusions between NUTM1 and members of the MAD (MAX dimerization) gene family of MYC antagonists. Here we describe a small round cell malignancy from the gastro-esophageal junction with a previously unreported fusion between NUTM1 and the MAD family member MXI1. In contrast to NCs, the MXI1-NUTM1 tumor did not show squamous differentiation and did not express MYC, TP63 or SOX2, genes known to be targets of BRD-NUTM1 proteins and critical for NC oncogenesis. Transcriptome analysis showed paradoxical enrichment of MYC target genes in the MXI1-NUTM1 tumor despite the lack of MYC expression. When expressed in vitro MXI1-NUTM1 partially phenocopied MYC, enhancing cell proliferation and cooperating with oncogenic HRAS to produce anchorage-independent cell growth. These data provide evidence that MAD family members, which are normally repressors of MYC activity, can be converted into MYC-like mimics by fusion to NUTM1. The pathological features and novel oncogenic mechanism of the MXI1-NUTM1 tumor show that identification of NUTM1 fusion partners can be important for accurate diagnostic classification of some NRN subtypes, and potentially may guide therapeutic options.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Esophageal Neoplasms/genetics , Esophagogastric Junction/pathology , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Stomach Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Fatal Outcome , Female , Humans , Middle Aged , Oncogene Proteins, Fusion , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Transcriptome
2.
Int J Surg Pathol ; 28(5): 553-562, 2020 Aug.
Article in English | MEDLINE | ID: mdl-31931637

ABSTRACT

The transcription factor GLI1 is a critical effector of the sonic hedgehog pathway. Gene fusions that activate GLI1 have recently been reported in several tumor types including gastroblastoma, plexiform fibromyxoma, a subset of pericytomas, and other soft tissue tumors. These tumors arise in a wide variety of anatomical origins and have variable malignant potentials, morphologies, and immunohistochemistry profiles. In this case report, we describe a malignant tumor from the jejunum with a MALAT1-GLI1 gene fusion that expressed a truncated constitutively active GLI1 protein and GLI1 targets that were detectable by immunohistochemistry. The tumor showed high-grade epithelioid and spindle cell morphology, strongly expressed CD56, and focally expressed other neuroendocrine markers and cytokeratins, but not S100 protein or SMA. The tumor recurred multiple times in liver, soft tissue, and lung over the course of 26 years, the longest reported follow-up for a GLI1 fusion-associated tumor. These metastatic tumors were also composed of epithelioid and spindle cells, but showed lower morphological grade than the primary tumor. The metastatic tumors resembled the recently reported "malignant epithelioid neoplasms with GLI1 rearrangements." The tumor also had a relatively high tumor mutation burden for a sarcoma. This case report expands the sites of origin for GLI1 rearranged neoplasms and shows that despite being associated with high-grade morphology, these malignancies can be associated with very long-term survival.


Subject(s)
Jejunum/pathology , RNA, Long Noncoding/genetics , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/pathology , Zinc Finger Protein GLI1/genetics , Humans , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Oncogene Proteins, Fusion/genetics , Soft Tissue Neoplasms/mortality
3.
Biotechniques ; 68(1): 48-51, 2020 01.
Article in English | MEDLINE | ID: mdl-31825238

ABSTRACT

Tumor DNA sequencing results can have important clinical implications. However, its use is often limited by low DNA input, owing to small tumor biopsy size. To help overcome this limitation we have developed a simple improvement to a commonly used next-generation sequencing (NGS) capture-based library preparation method using formalin-fixed paraffin-embedded-derived tumor DNA. By using on-bead PCR for pre-capture library generation we show that library yields are dramatically increased, resulting in decreased sample failure rates. Improved yields allowed for a reduction in PCR cycles, which translated into improved sequencing parameters without affecting variant calling. This methodology should be applicable to any NGS system in which input DNA is a limiting factor.


Subject(s)
Gene Library , High-Throughput Nucleotide Sequencing/methods , Polymerase Chain Reaction/methods , Humans , Neoplasms/genetics , Polymerase Chain Reaction/instrumentation
4.
J Clin Invest ; 129(5): 1940-1945, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30835257

ABSTRACT

BRAF and CRAF are critical components of the MAPK signaling pathway which is activated in many cancer types. In approximately 1% of melanomas, BRAF or CRAF are activated through structural arrangements. We describe here a metastatic melanoma with a GOLGA4-RAF1 fusion and pathogenic variants in CTNNB1 and CDKN2A. Anti-CTLA4/anti-PD1 combination immunotherapy failed to control tumor progression. In the absence of other actionable variants the patient was administered MEK inhibitor therapy on the basis of its potential action against RAF1 fusions. This resulted in a profound and clinically significant response. We demonstrated that GOLGA4-RAF1 expression was associated with ERK activation, elevated expression of the RAS/RAF downstream co-effector ETV5, and a high Ki67 index. These findings provide a rationale for the dramatic response to targeted therapy. This study shows that thorough molecular characterization of treatment-resistant cancers can identify therapeutic targets and personalize management, leading to improved patient outcomes.


Subject(s)
Autoantigens/genetics , MAP Kinase Kinase 1/antagonists & inhibitors , Melanoma/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-raf/genetics , Skin Neoplasms/genetics , Aged , Alleles , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fluorodeoxyglucose F18/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasm Metastasis , Oncogene Proteins, Fusion/metabolism , Positron-Emission Tomography , beta Catenin/metabolism
5.
NPJ Precis Oncol ; 2: 9, 2018.
Article in English | MEDLINE | ID: mdl-29872718

ABSTRACT

Succinate dehydrogenase (SDH)-deficient renal cell carcinoma (RCC) is a rare RCC subtype that is caused by biallelic mutation of one of the four subunits of the SDH complex (SDHA, B, C, and D) and results in inactivation of the SDH enzyme. Here we describe a case of genetically characterized SDH-deficient RCC caused by biallelic (germline plus somatic) SDHA mutations. SDHA pathogenic variants were detected using comprehensive genomic profiling and SDH absence was subsequently confirmed by immunohistochemistry. Very little is known regarding the genomic context of SDH-deficient RCC. Interestingly we found genomic amplifications commonly observed in RCC but there was an absence of additional variants in common cancer driver genes. Prior to genetic testing a PD-1 inhibitor treatment was administered. However, following the genetic results a succession of tyrosine kinase inhibitors were administered as targeted treatment options and we highlight how the genetic results provide a rationale for their effectiveness. We also describe how the genetic results benefited the patient by empowering him to adopt dietary and lifestyle changes in accordance with knowledge of the mechanisms of SDH-related tumorigenesis.

6.
Mod Pathol ; 30(7): 952-963, 2017 07.
Article in English | MEDLINE | ID: mdl-28338653

ABSTRACT

The spectrum of genomic alterations in ductal carcinoma in situ (DCIS) is relatively unexplored, but is likely to provide useful insights into its biology, its progression to invasive carcinoma and the risk of recurrence. DCIS (n=20) with a range of phenotypes was assessed by massively parallel sequencing for mutations and copy number alterations and variants validated by Sanger sequencing. PIK3CA mutations were identified in 11/20 (55%), TP53 mutations in 6/20 (30%), and GATA3 mutations in 9/20 (45%). Screening an additional 91 cases for GATA3 mutations identified a final frequency of 27% (30/111), with a high proportion of missense variants (8/30). TP53 mutations were exclusive to high grade DCIS and more frequent in PR-negative tumors compared with PR-positive tumors (P=0.037). TP53 mutant tumors also had a significantly higher fraction of the genome altered by copy number than wild-type tumors (P=0.005), including a significant positive association with amplification or gain of ERBB2 (P<0.05). The association between TP53 mutation and ERBB2 amplification was confirmed in a wider DCIS cohort using p53 immunohistochemistry as a surrogate marker for TP53 mutations (P=0.03). RUNX1 mutations and MAP2K4 copy number loss were novel findings in DCIS. Frequent copy number alterations included gains on 1q, 8q, 17q, and 20q and losses on 8p, 11q, 16q, and 17p. Patterns of genomic alterations observed in DCIS were similar to those previously reported for invasive breast cancers, with all DCIS having at least one bona fide breast cancer driver event. However, an increase in GATA3 mutations and fewer copy number changes were noted in DCIS compared with invasive carcinomas. The role of such alterations as prognostic and predictive biomarkers in DCIS is an avenue for further investigation.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Intraductal, Noninfiltrating/genetics , Mutation , Adult , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Class I Phosphatidylinositol 3-Kinases/genetics , DNA Copy Number Variations , Female , GATA3 Transcription Factor/genetics , Humans , Middle Aged , Receptor, ErbB-2/genetics , Tumor Suppressor Protein p53/genetics
7.
Mod Pathol ; 28(9): 1174-84, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26321097

ABSTRACT

Ductal carcinoma in situ (DCIS) is a non-obligate precursor of invasive breast cancer and a frequent mammographic finding requiring treatment. Up to 25% of DCIS can recur and half of recurrences are invasive, but there are no reliable biomarkers for recurrence. We hypothesised that copy number aberrations could predict likelihood of recurrence. We analysed a cohort of pure DCIS cases treated only with wide local excision for genome-wide copy number and loss of heterozygosity using Affymetrix OncoScan MIP arrays. Cases included those without recurrence within 7 years (n = 25) and with recurrence between 1 and 5 years after diagnosis (n = 15). Pure DCIS were broadly similar in copy number changes compared with invasive breast cancer, with the consistent exception of a greater frequency of ERBB2 amplification in DCIS. There were no significant differences in age or ER status between the cases with a recurrence vs those without. Overall, the DCIS cases with recurrence had more copy number events than the DCIS without recurrence. The increased copy number appeared non-random with several genomic regions showing an increase in frequency in recurrent cases, including 20 q gain, ERBB2 amplification and 15q loss. Copy number changes may provide prognostic information for DCIS recurrence, but validation in additional cohorts is required.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Intraductal, Noninfiltrating/genetics , Neoplasm Recurrence, Local/genetics , Aged , Female , Gene Dosage , Humans , Immunohistochemistry , Middle Aged , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction
8.
BMC Med Genomics ; 8: 45, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26231170

ABSTRACT

BACKGROUND: Loss of heterozygosity (LOH) is a common genetic event in cancer development, and is known to be involved in the somatic loss of wild-type alleles in many inherited cancer syndromes. The wider involvement of LOH in cancer is assumed to relate to unmasking a somatically mutated tumour suppressor gene through loss of the wild type allele. METHODS: We analysed 86 ovarian carcinomas for mutations in 980 genes selected on the basis of their location in common regions of LOH. RESULTS: We identified 36 significantly mutated genes, but these could only partly account for the quanta of LOH in the samples. Using our own and TCGA data we then evaluated five possible models to explain the selection for non-random accumulation of LOH in ovarian cancer genomes: 1. Classic two-hit hypothesis: high frequency biallelic genetic inactivation of tumour suppressor genes. 2. Epigenetic two-hit hypothesis: biallelic inactivation through methylation and LOH. 3. Multiple alternate-gene biallelic inactivation: low frequency gene disruption. 4. Haplo-insufficiency: Single copy gene disruption. 5. Modified two-hit hypothesis: reduction to homozygosity of low penetrance germline predisposition alleles. We determined that while high-frequency biallelic gene inactivation under model 1 is rare, regions of LOH (particularly copy-number neutral LOH) are enriched for deleterious mutations and increased promoter methylation, while copy-number loss LOH regions are likely to contain under-expressed genes suggestive of haploinsufficiency. Reduction to homozygosity of cancer predisposition SNPs may also play a minor role. CONCLUSION: It is likely that selection for regions of LOH depends on its effect on multiple genes. Selection for copy number neutral LOH may better fit the classic two-hit model whereas selection for copy number loss may be attributed to its effect on multi-gene haploinsufficiency. LOH mapping alone is unlikely to be successful in identifying novel tumour suppressor genes; a combined approach may be more effective.


Subject(s)
Loss of Heterozygosity , Ovarian Neoplasms/genetics , Female , Gene Dosage/genetics , Genes, Neoplasm/genetics , Genes, Tumor Suppressor , Genomics , Haploinsufficiency/genetics , Humans
9.
Genome Med ; 5(9): 90, 2013.
Article in English | MEDLINE | ID: mdl-24073752

ABSTRACT

Differentiating true somatic mutations from artifacts in massively parallel sequencing data is an immense challenge. To develop methods for optimal somatic mutation detection and to identify factors influencing somatic mutation prediction accuracy, we validated predictions from three somatic mutation detection algorithms, MuTect, JointSNVMix2 and SomaticSniper, by Sanger sequencing. Full consensus predictions had a validation rate of >98%, but some partial consensus predictions validated too. In cases of partial consensus, read depth and mapping quality data, along with additional prediction methods, aided in removing inaccurate predictions. Our consensus approach is fast, flexible and provides a high-confidence list of putative somatic mutations.

10.
PLoS Genet ; 8(9): e1002894, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23028338

ABSTRACT

Despite intensive efforts using linkage and candidate gene approaches, the genetic etiology for the majority of families with a multi-generational breast cancer predisposition is unknown. In this study, we used whole-exome sequencing of thirty-three individuals from 15 breast cancer families to identify potential predisposing genes. Our analysis identified families with heterozygous, deleterious mutations in the DNA repair genes FANCC and BLM, which are responsible for the autosomal recessive disorders Fanconi Anemia and Bloom syndrome. In total, screening of all exons in these genes in 438 breast cancer families identified three with truncating mutations in FANCC and two with truncating mutations in BLM. Additional screening of FANCC mutation hotspot exons identified one pathogenic mutation among an additional 957 breast cancer families. Importantly, none of the deleterious mutations were identified among 464 healthy controls and are not reported in the 1,000 Genomes data. Given the rarity of Fanconi Anemia and Bloom syndrome disorders among Caucasian populations, the finding of multiple deleterious mutations in these critical DNA repair genes among high-risk breast cancer families is intriguing and suggestive of a predisposing role. Our data demonstrate the utility of intra-family exome-sequencing approaches to uncover cancer predisposition genes, but highlight the major challenge of definitively validating candidates where the incidence of sporadic disease is high, germline mutations are not fully penetrant, and individual predisposition genes may only account for a tiny proportion of breast cancer families.


Subject(s)
Breast Neoplasms/genetics , Fanconi Anemia Complementation Group C Protein/genetics , RecQ Helicases/genetics , Sequence Deletion/genetics , Alleles , Exome/genetics , Exons , Fanconi Anemia Complementation Group C Protein/metabolism , Female , Genetic Predisposition to Disease , Genome, Human , Humans , Pedigree , Polymorphism, Genetic , RecQ Helicases/metabolism , Sequence Analysis, DNA
11.
Breast Cancer Res Treat ; 134(2): 543-7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22580995

ABSTRACT

KLLN is a p53 target gene with DNA binding function and represents a highly plausible candidate breast cancer predisposition gene. We screened for predisposing variants in 860 high-risk breast cancer families using high resolution melt analysis. A germline c.339_340delAG variant predicted to cause premature termination of the protein after 57 alternative amino acid residues was identified in 3/860 families who tested negative for BRCA1 and BRCA2 mutations and in 1/84 sporadic breast cancer cases. However, the variant was also detected in 2/182 families with known BRCA1 or BRCA2 mutations and in 2/464 non-cancer controls. Furthermore, loss of the mutant allele was detected in 2/2 breast tumors. Our data suggest that pathogenic mutations in KLLN are rare in breast cancer families and the c.339_340delAG variant does not represent a high-penetrance breast cancer risk allele.


Subject(s)
Breast Neoplasms/genetics , Genetic Predisposition to Disease , Penetrance , Tumor Suppressor Proteins/genetics , Adult , Base Sequence , Case-Control Studies , Cohort Studies , DNA Mutational Analysis , Female , Gene Frequency , Genetic Association Studies , Germ-Line Mutation , Humans , Loss of Heterozygosity , Molecular Sequence Data , Sequence Deletion
12.
PLoS One ; 7(4): e35805, 2012.
Article in English | MEDLINE | ID: mdl-22536442

ABSTRACT

MicroRNAs are key regulators of gene expression and have been shown to have altered expression in a variety of cancer types, including epithelial ovarian cancer. MiRNA function is most often achieved through binding to the 3'-untranslated region of the target protein coding gene. Mutation screening using massively-parallel sequencing of 712 miRNA genes in 86 ovarian cancer cases identified only 5 mutated miRNA genes, each in a different case. One mutation was located in the mature miRNA, and three mutations were predicted to alter the secondary structure of the miRNA transcript. Screening of the 3'-untranslated region of 18 candidate cancer genes identified one mutation in each of AKT2, EGFR, ERRB2 and CTNNB1. The functional effect of these mutations is unclear, as expression data available for AKT2 and EGFR showed no increase in gene transcript. Mutations in miRNA genes and 3'-untranslated regions are thus uncommon in ovarian cancer.


Subject(s)
3' Untranslated Regions , MicroRNAs/genetics , Mutation , Ovarian Neoplasms/genetics , Binding Sites/genetics , DNA Mutational Analysis , ErbB Receptors/genetics , Female , Genetic Association Studies , Humans , Proto-Oncogene Proteins c-akt/genetics , Receptor, ErbB-2/genetics , beta Catenin/genetics
13.
Hum Mutat ; 33(1): 95-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21990120

ABSTRACT

There is strong evidence that overtly inactivating mutations in RAD51C predispose to hereditary breast and ovarian cancer but the prevalence of such mutations, and whether they are associated with a particular clinical phenotype, remains unclear. Resolving these questions has important implications for the implementation of RAD51C into routine clinical genetic testing. Consequently, we have performed a large RAD51C mutation screen of hereditary breast and ovarian cancer families, and the first study of unselected patients diagnosed with ovarian cancer. Our data confirm a consistent but low frequency (2/335 families) of inactivating RAD51C mutations among families with a history of both breast and ovarian cancer and an absence of mutations among breast cancer only families (0/1,053 families). Our data also provide support for the designation of the missense variant p.Gly264Ser as a moderate penetrance allele.


Subject(s)
Breast Neoplasms/genetics , DNA-Binding Proteins/genetics , Mutation, Missense , Ovarian Neoplasms/genetics , Adult , Comorbidity , DNA Mutational Analysis , Female , Gene Frequency , Genetic Predisposition to Disease , Genetic Testing , Germ-Line Mutation , Humans , Middle Aged , Pedigree , Penetrance , Risk Factors
14.
BMC Cancer ; 11: 173, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21575258

ABSTRACT

BACKGROUND: MAP2K4 is a putative tumor and metastasis suppressor gene frequently found to be deleted in various cancer types. We aimed to conduct a comprehensive analysis of this gene to assess its involvement in ovarian cancer. METHODS: We screened for mutations in MAP2K4 using High Resolution Melt analysis of 149 primary ovarian tumors and methylation at the promoter using Methylation-Specific Single-Stranded Conformation Polymorphism analysis of 39 tumors. We also considered the clinical impact of changes in MAP2K4 using publicly available expression and copy number array data. Finally, we used siRNA to measure the effect of reducing MAP2K4 expression in cell lines. RESULTS: In addition to 4 previously detected homozygous deletions, we identified a homozygous 16 bp truncating deletion and a heterozygous 4 bp deletion, each in one ovarian tumor. No promoter methylation was detected. The frequency of MAP2K4 homozygous inactivation was 5.6% overall, and 9.8% in high-grade serous cases. Hemizygous deletion of MAP2K4 was observed in 38% of samples. There were significant correlations of copy number and expression in three microarray data sets. There was a significant correlation between MAP2K4 expression and overall survival in one expression array data set, but this was not confirmed in an independent set. Treatment of JAM and HOSE6.3 cell lines with MAP2K4 siRNA showed some reduction in proliferation. CONCLUSIONS: MAP2K4 is targeted by genetic inactivation in ovarian cancer and restricted to high grade serous and endometrioid carcinomas in our cohort.


Subject(s)
MAP Kinase Kinase 4/genetics , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Adult , Aged , Aged, 80 and over , Base Sequence , Cell Proliferation , DNA Methylation/genetics , Female , Gene Dosage/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Genotype , Humans , Middle Aged , Molecular Sequence Data , Mutation/genetics , Ovarian Neoplasms/mortality , Promoter Regions, Genetic/genetics , Survival Analysis
15.
Breast Cancer Res Treat ; 119(2): 491-6, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19189213

ABSTRACT

Heterozygous somatic mutations of the transcription factor, GATA-3, have recently been reported in approximately 5% breast of tumors unselected for family history. We sequenced the GATA-3 gene in 55 breast tumors from women with familial breast cancer, and found seven heterozygous somatic mutations, all in non-BRCA1/2 cases in which the frequency was 22%. In contrast, we found mutations of GATA-3 in only 4% of 81 sporadic tumors analysed. It is possible that GATA3 mutations occur earlier in the evolution of BRCAx tumors, compared to BRCA1, BRCA2 or sporadic tumors, and are therefore easier to detect by direct sequencing in the presence of some stromal contamination.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , Breast Neoplasms/genetics , GATA3 Transcription Factor/genetics , Mutation , Apoptosis Regulatory Proteins , Breast Neoplasms/pathology , DNA Mutational Analysis , Female , Gene Expression Regulation, Neoplastic , Gene Frequency , Genetic Predisposition to Disease , Heterozygote , Humans , Pedigree , Risk Assessment , Risk Factors
16.
Clin Cancer Res ; 14(22): 7246-50, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-19010840

ABSTRACT

PURPOSE: There is accumulating evidence that microRNAs may function like classic tumor suppressor genes but little is known about their mechanism of inactivation in cancer cells. We investigated whether somatic mutations are a common mechanism of inactivation of microRNA genes in ovarian cancer. EXPERIMENTAL DESIGN: Ten cancer-implicated microRNA genes were analyzed for somatic mutations in 90 ovarian epithelial cancers and matching normal DNA. High-resolution melt analysis and bidirectional sequencing was used to detect sequence variations. RESULTS: High-resolution melt analysis and direct sequencing did not identify any somatic mutations but did reveal numerous novel and previously reported germ line base substitutions, deletions, and insertions surrounding the mature microRNA sequences. The majority of variants were detected in the same proportion of non-cancer control individuals suggesting that they do not represent ovarian cancer-predisposing alleles. CONCLUSION: The absence of somatic mutations in any of the 10 cancer-implicated microRNAs in our large cohort of ovarian tumors suggests that this may be an uncommon mechanism of inactivation of microRNAs in ovarian cancer.


Subject(s)
MicroRNAs/genetics , Ovarian Neoplasms/genetics , DNA, Neoplasm/genetics , Female , Humans , Mutation , Polymerase Chain Reaction
17.
Neoplasia ; 10(11): 1253-8, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18953434

ABSTRACT

Chromodomain, helicase, DNA binding 5 (CHD5) is a member of a subclass of the chromatin remodeling Swi/Snf proteins and has recently been proposed as a tumor suppressor in a diverse range of human cancers. We analyzed all 41 coding exons of CHD5 for somatic mutations in 123 primary ovarian cancers as well as 60 primary breast cancers using high-resolution melt analysis. We also examined methylation of the CHD5 promoter in 48 ovarian cancer samples by methylation-specific single-stranded conformation polymorphism and bisulfite sequencing. In contrast to previous studies, no mutations were identified in the breast cancers, but somatic heterozygous missense mutations were identified in 3 of 123 ovarian cancers. We identified promoter methylation in 3 of 45 samples with normal CHD5 and in 2 of 3 samples with CHD5 mutation, suggesting these tumors may have biallelic inactivation of CHD5. Hemizygous copy number loss at CHD5 occurred in 6 of 85 samples as assessed by single nucleotide polymorphism array. Tumors with CHD5 mutation or methylation were more likely to have mutation of KRAS or BRAF (P = .04). The aggregate frequency of CHD5 haploinsufficiency or inactivation is 16.2% in ovarian cancer. Thus, CHD5 may play a role as a tumor suppressor gene in ovarian cancer; however, it is likely that there is another target of the frequent copy number neutral loss of heterozygosity observed at 1p36.


Subject(s)
DNA Helicases/genetics , DNA Methylation , Mutation , Nerve Tissue Proteins/genetics , Ovarian Neoplasms/genetics , CpG Islands , DNA Mutational Analysis , Female , Gene Dosage , Genes, Tumor Suppressor , Humans , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/metabolism , Polymorphism, Genetic , Polymorphism, Single-Stranded Conformational , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras) , Reverse Transcriptase Polymerase Chain Reaction , ras Proteins/genetics , ras Proteins/metabolism
18.
Breast Cancer Res Treat ; 112(2): 351-2, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18095152

ABSTRACT

Mutations in BRCA1 predispose to breast cancer. CTIP interacts with BRCA1 and so could also be associated with increased risk. We screened CTIP for germline mutations in 210 probands of breast cancer families including 129 families with no mutations in BRCA1 or BRCA2. No coding variants were detected in CTIP, therefore, it is unlikely to be involved in breast cancer risk.


Subject(s)
BRCA1 Protein/genetics , BRCA1 Protein/physiology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carrier Proteins/genetics , Carrier Proteins/physiology , Genes, BRCA1 , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Australia , Breast Neoplasms/ethnology , DNA Mutational Analysis , Endodeoxyribonucleases , Family Health , Female , Genetic Predisposition to Disease , Humans , Male , Mass Screening/methods , Microsatellite Repeats , Risk
19.
Breast Cancer Res Treat ; 111(3): 505-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-17972171

ABSTRACT

Several studies in various populations have suggested that non-synonymous BARD1 variants are associated with increased breast cancer risk. Using DHPLC analysis we screened the coding region of BARD1 for variants in 210 probands of breast cancer families including 129 families with no mutations in BRCA1 or BRCA2. These families were ascertained in Australia through the Kathleen Cunningham Foundation Consortium for Research into Familial Breast Cancer (kConFab). Nine coding variants were detected among the kConFab families, including two novel variants (Thr598Ile and Ile692Thr). The frequency of five of these variants were evaluated in 258 non-cancer controls and 401 women with sporadic breast cancer. Three variants (1139del21, G1756C and A2285G) were detected in all three groups at a similar frequency suggesting that these do not represent BRCAX candidates. Two variants (Thr598Ile and Ile692Thr) were not detected in any of the 659 sporadic breast cancer cases and controls and were assessed for segregation with breast cancer in the families of the probands. However, neither variant was identified in any other breast cancer case in either family suggesting that these variants are non-pathogenic polymorphisms. We have found no evidence to support involvement of BARD1 in familial breast cancer risk in the Australian population. In addition, three variants previously reported to be pathogenic in other populations are likely to represent benign polymorphisms and therefore we conclude that BARD1 is unlikely to represent a high-penetrance breast cancer susceptibility gene.


Subject(s)
Breast Neoplasms/genetics , Polymorphism, Genetic , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Australia , Breast Neoplasms/pathology , Case-Control Studies , Female , Gene Frequency , Genetic Predisposition to Disease , Humans , Pedigree , Risk Assessment , Risk Factors
20.
Clin Cancer Res ; 13(16): 4731-9, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17699850

ABSTRACT

PURPOSE: Genetic changes in sporadic ovarian cancer are relatively poorly characterized compared with other tumor types. We have evaluated the use of high-resolution whole genome arrays for the genetic profiling of epithelial ovarian cancer. EXPERIMENTAL DESIGN: We have evaluated 31 primary ovarian cancers and matched normal DNA for loss of heterozygosity and copy number alterations using 500 K single nucleotide polymorphism arrays. RESULTS: In addition to identifying the expected large-scale genomic copy number changes, >380 small regions of copy number gain or loss (<500 kb) were identified among the 31 tumors, including 33 regions of high-level gain (>5 copies) and 27 homozygous deletions. The existence of such a high frequency of small regions exhibiting copy number alterations had not been previously suspected because earlier genomic array platforms lacked comparable resolution. Interestingly, many of these regions harbor known cancer genes. For example, one tumor harbored a 350-kb high-level amplification centered on FGFR1 and three tumors showed regions of homozygous loss 109 to 216 kb in size involving the RB1 tumor suppressor gene only. CONCLUSIONS: These data suggest that novel cancer genes may be located within the other identified small regions of copy number alteration. Analysis of the number of copy number breakpoints and the distribution of the small regions of copy number change indicate high levels of structural chromosomal genetic instability in ovarian cancer.


Subject(s)
Chromosome Aberrations , Gene Dosage , Neoplasms, Glandular and Epithelial/genetics , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/genetics , Polymorphism, Single Nucleotide , Allelic Imbalance , Chromosomes, Human, Pair 9 , Female , Gene Amplification , Gene Deletion , Humans , MAP Kinase Kinase 4/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...