Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Ann Oncol ; 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38908482

ABSTRACT

BACKGROUND: Hormone receptor expression is a known positive prognostic and predictive factor in breast cancer; however, limited evidence exists on its impact on prognosis of young patients harboring BRCA pathogenic variant (PV). PATIENTS AND METHODS: This international, multicenter, retrospective cohort study included young patients (≤40 years) diagnosed with invasive breast cancer and harboring germline PV in BRCA genes. We investigated the impact of hormone receptor status on clinical behavior and outcomes of breast cancer. Outcomes of interest (disease-free survival [DFS], breast cancer specific survival [BCSS] and overall survival [OS]) were first investigated according to hormone receptors expression (positive vs. negative), and then according to breast cancer subtype (luminal A-like vs. luminal B-like vs. triple-negative vs. HER2-positive breast cancer). RESULTS: From 78 centers worldwide, 4,709 BRCA carriers were included, of whom 2,143 (45.5%) had hormone receptor-positive and 2,566 (54.5%) hormone receptor-negative breast cancer. Median follow-up was 7.9 years. The rate of distant recurrences was higher in patients with hormone receptor-positive disease (13.1% vs. 9.6%, p<0.001), while the rate of second primary breast cancer was lower (9.1% vs. 14.7%, p<0.001) compared to patients with hormone receptor-negative disease. The 8-years DFS was 65.8% and 63.4% in patients with hormone receptor-positive and negative disease, respectively. The hazard ratio of hormone receptor-positive vs. negative disease changed over time for DFS, BCSS, and OS (p<0.05 for interactions of hormone receptor status and survival time). Patients with luminal A-like breast cancer had the worst long-term prognosis in terms of DFS compared to all the other subgroups (8-years DFS: 60.8% in luminal A-like vs. 63.5% in triple-negative vs. 65.5% in HER2-positive and 69.7% in luminal B-like subtype). CONCLUSIONS: In young BRCA carriers, differences in recurrence pattern and second primary breast cancer among hormone receptor-positive vs. negative disease warrants consideration in counseling patients on treatment, follow-up, and risk-reducing surgery.

2.
Ann Oncol ; 26(10): 2057-65, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26153499

ABSTRACT

BACKGROUND: Increasing use of BRCA1/2 testing for tailoring cancer treatment and extension of testing to tumour tissue for somatic mutation is moving BRCA1/2 mutation screening from a primarily prevention arena delivered by specialist genetic services into mainstream oncology practice. A considerable number of gene tests will identify rare variants where clinical significance cannot be inferred from sequence information alone. The proportion of variants of uncertain clinical significance (VUS) is likely to grow with lower thresholds for testing and laboratory providers with less experience of BRCA. Most VUS will not be associated with a high risk of cancer but a misinterpreted VUS has the potential to lead to mismanagement of both the patient and their relatives. DESIGN: Members of the Clinical Working Group of ENIGMA (Evidence-based Network for the Interpretation of Germline Mutant Alleles) global consortium (www.enigmaconsortium.org) observed wide variation in practices in reporting, disclosure and clinical management of patients with a VUS. Examples from current clinical practice are presented and discussed to illustrate potential pitfalls, explore factors contributing to misinterpretation, and propose approaches to improving clarity. RESULTS AND CONCLUSION: Clinicians, patients and their relatives would all benefit from an improved level of genetic literacy. Genetic laboratories working with clinical geneticists need to agree on a clinically clear and uniform format for reporting BRCA test results to non-geneticists. An international consortium of experts, collecting and integrating all available lines of evidence and classifying variants according to an internationally recognized system, will facilitate reclassification of variants for clinical use.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , Breast Neoplasms/diagnosis , Genetic Testing/standards , Genetic Variation/genetics , Mutation/genetics , Ovarian Neoplasms/diagnosis , Breast Neoplasms/genetics , Data Interpretation, Statistical , Female , Genetic Predisposition to Disease , Humans , Ovarian Neoplasms/genetics , Practice Guidelines as Topic , Prognosis , Risk Factors
3.
Clin Genet ; 88(6): 523-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25640009

ABSTRACT

Studies indicate variant of uncertain significance (VUS) results are challenging for genetic counselors and patients, often resulting in negative patient outcomes. Genetic counselors' current practices regarding VUS are unknown. This study utilized a national survey of genetic counselors (n = 932) to examine current practices and confidence related to disclosing BRCA VUS results and reclassification information. For participants (n = 398), descriptive statistics were calculated regarding patient demographic characteristics, practices and confidence, and cross tabulation was used to identify participant's actions when receiving a reclassified VUS. Upon receiving a BRCA VUS report, the majority reported providing patients with information about the frequency with which their VUS was seen and patient ancestry, but a minority discussed DNA banking. Most were confident in their understanding of, and ability to explain, VUS results to patients, but felt less confident about achieving high levels of patient understanding. Upon reclassification, the majority reported calling the patient and mailing the results, but when the reclassification was deleterious, the majority also met with the patient face-to-face. Given the lack of standard professional guidelines about informing patients of initial and reclassified VUS results, this overview provides important insight into genetic counselors' current practices and confidence.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , Genetic Counseling/methods , Genetic Testing/methods , Genetic Variation , Adult , Aged , Female , Genetic Counseling/psychology , Genetic Predisposition to Disease/genetics , Health Surveys/methods , Health Surveys/statistics & numerical data , Humans , Male , Middle Aged , Mutation , Professional-Patient Relations , Truth Disclosure , Uncertainty , Young Adult
4.
Br J Cancer ; 107(5): 800-7, 2012 Aug 21.
Article in English | MEDLINE | ID: mdl-22850554

ABSTRACT

BACKGROUND: Predict (www.predict.nhs.uk) is an online, breast cancer prognostication and treatment benefit tool. The aim of this study was to incorporate the prognostic effect of HER2 status in a new version (Predict+), and to compare its performance with the original Predict and Adjuvant!. METHODS: The prognostic effect of HER2 status was based on an analysis of data from 10 179 breast cancer patients from 14 studies in the Breast Cancer Association Consortium. The hazard ratio estimates were incorporated into Predict. The validation study was based on 1653 patients with early-stage invasive breast cancer identified from the British Columbia Breast Cancer Outcomes Unit. Predicted overall survival (OS) and breast cancer-specific survival (BCSS) for Predict+, Predict and Adjuvant! were compared with observed outcomes. RESULTS: All three models performed well for both OS and BCSS. Both Predict models provided better BCSS estimates than Adjuvant!. In the subset of patients with HER2-positive tumours, Predict+ performed substantially better than the other two models for both OS and BCSS. CONCLUSION: Predict+ is the first clinical breast cancer prognostication tool that includes tumour HER2 status. Use of the model might lead to more accurate absolute treatment benefit predictions for individual patients.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Models, Statistical , Receptor, ErbB-2/biosynthesis , Adult , Aged , Cohort Studies , Female , Humans , Middle Aged , Prognosis , Proportional Hazards Models , Reproducibility of Results , Young Adult
5.
Br J Cancer ; 104(8): 1356-61, 2011 Apr 12.
Article in English | MEDLINE | ID: mdl-21427728

ABSTRACT

BACKGROUND: Single-nucleotide polymorphisms (SNPs) in genes involved in DNA repair are good candidates to be tested as phenotypic modifiers for carriers of mutations in the high-risk susceptibility genes BRCA1 and BRCA2. The base excision repair (BER) pathway could be particularly interesting given the relation of synthetic lethality that exists between one of the components of the pathway, PARP1, and both BRCA1 and BRCA2. In this study, we have evaluated the XRCC1 gene that participates in the BER pathway, as phenotypic modifier of BRCA1 and BRCA2. METHODS: Three common SNPs in the gene, c.-77C>T (rs3213245) p.Arg280His (rs25489) and p.Gln399Arg (rs25487) were analysed in a series of 701 BRCA1 and 576 BRCA2 mutation carriers. RESULTS: An association was observed between p.Arg280His-rs25489 and breast cancer risk for BRCA2 mutation carriers, with rare homozygotes at increased risk relative to common homozygotes (hazard ratio: 22.3, 95% confidence interval: 14.3-34, P<0.001). This association was further tested in a second series of 4480 BRCA1 and 3016 BRCA2 mutation carriers from the Consortium of Investigators of Modifiers of BRCA1 and BRCA2. CONCLUSIONS AND INTERPRETATION: No evidence of association was found when the larger series was analysed which lead us to conclude that none of the three SNPs are significant modifiers of breast cancer risk for mutation carriers.


Subject(s)
Breast Neoplasms/genetics , Carcinoma/genetics , DNA-Binding Proteins/physiology , Epistasis, Genetic/physiology , Genes, BRCA1 , Genes, BRCA2 , Adolescent , Adult , Aged , Aged, 80 and over , Breast Neoplasms/epidemiology , Carcinoma/epidemiology , DNA-Binding Proteins/genetics , Female , Focus Groups , Genes, BRCA1/physiology , Genes, BRCA2/physiology , Genetic Predisposition to Disease , Heterozygote , Humans , Middle Aged , Phenotype , Polymorphism, Single Nucleotide , X-ray Repair Cross Complementing Protein 1 , Young Adult
6.
Oncogene ; 30(34): 3705-15, 2011 Aug 25.
Article in English | MEDLINE | ID: mdl-21441955

ABSTRACT

Glycogen synthase kinase-3 beta (GSK-3ß) is overexpressed in a number of human malignancies and has been shown to contribute to tumor cell proliferation and survival. Although regulation of GSK-3ß activity has been extensively studied, the mechanisms governing GSK-3ß gene expression are still unknown. Using pancreatic cancer as a model, we find that constitutively active Ras signaling increases GSK-3ß gene expression via the canonical mitogen-activated protein kinase signaling pathway. Analysis of the mechanism revealed that K-Ras regulates the expression of this kinase through two highly conserved E-twenty six (ETS) binding elements within the proximal region. Furthermore, we demonstrate that mutant K-Ras enhances ETS2 loading onto the promoter, and ETS requires its transcriptional activity to increase GSK-3ß gene transcription in pancreatic cancer cells. Lastly, we show that ETS2 cooperates with p300 histone acetyltransferase to remodel chromatin and promote GSK-3ß expression. Taken together, these results provide a general mechanism for increased expression of GSK-3ß in pancreatic cancer and perhaps other cancers, where Ras signaling is deregulated.


Subject(s)
Genes, ras , Glycogen Synthase Kinase 3/genetics , Mutation , Pancreatic Neoplasms/genetics , RNA-Binding Protein EWS/genetics , p300-CBP Transcription Factors/genetics , Animals , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Immunohistochemistry , MAP Kinase Signaling System , Mice , Mice, Transgenic , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic , Transcription, Genetic
7.
Breast Cancer Res Treat ; 125(1): 221-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20508983

ABSTRACT

Centrosome amplification has been detected in premalignant lesions and in situ tumors in the breast and in over 70% of invasive breast tumors, and has been associated with aneuploidy and tumor development. Based on these observations, the contribution of commonly inherited genetic variation in candidate genes related to centrosome structure and function to breast cancer risk was evaluated in an association study. Seven-hundred and 82 single nucleotide polymorphisms (SNPs) from 101 centrosomal genes were analyzed in 798 breast cancer cases and 843 controls from the Mayo Clinic Breast Cancer Study to assess the association between these SNPs (both individually and combined) and risk of breast cancer in this population. Eleven SNPs out of 782 from six genes displayed associations with breast cancer risk (P < 0.01). Haplotypes in five genes also displayed significant associations with risk. A two SNP combination of rs10145182 in NIN and rs2134808 in the TUBG1 locus (P-interaction = 0.00001), suggested SNPs in mediators of microtubule nucleation from the centrosome contribute to breast cancer. Evaluation of the simultaneous significance of all SNPs in the centrosome pathway suggested that the centrosome pathway is highly enriched (P = 4.76 × 10(-50)) for SNPs that are associated with breast cancer risk. Collections of weakly associated genetic variants in the centrosome pathway, rather than individual highly significantly associated SNPs, may account for a putative role for the centrosome pathway in predisposition to breast cancer.


Subject(s)
Breast Neoplasms/genetics , Centrosome/pathology , Polymorphism, Single Nucleotide , Breast Neoplasms/pathology , Case-Control Studies , Female , Gene Frequency , Genetic Predisposition to Disease , Haplotypes , Humans , Linkage Disequilibrium , Logistic Models , Minnesota , Odds Ratio , Risk Assessment , Risk Factors
8.
Breast Cancer Res Treat ; 119(2): 423-30, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19377877

ABSTRACT

The down-regulation of genes involved in normal cell division can cause aberrant mitoses and increased cell death. Surviving cells exhibit aneuploidy and/or polyploidy. Since mitotic disruption has been linked with tumor development and progression, alterations in the expression or activity of these mitotic regulators may contribute to breast tumor formation. We evaluated associations between common inherited variation in these genes and breast cancer risk. Two hundred and five tagging and candidate functional single nucleotide polymorphisms in 30 genes required for normal cell division were genotyped in 798 breast cancer cases and 843 controls from the Mayo Clinic breast cancer study. Two variants in EIF3A (rs10787899 and rs3824830; P < 0.01) and four variants in SART1 (rs660118, rs679581, rs754532, and rs735942; P(trend) < or = 0.02) were significantly associated with an altered risk of breast cancer along with single variants in RRM2, PSCD3, C11orf51, CDC16, SNW1, MFAP1, and CDC2 (P < 0.05). Variation in both SART1 (P = 0.009) and EIF3A (P = 0.02) was also significant at the gene level. Analyses suggested that SART1 SNPs rs660118 and rs679581 accounted for the majority of the association of that gene with breast cancer. The observed associations between breast cancer risk and genetic variation in the SART1 and EIF3A genes that are required for maintenance of normal mitosis suggest a direct role for these genes in the development of breast cancer.


Subject(s)
Antigens, Neoplasm/genetics , Breast Neoplasms/genetics , Eukaryotic Initiation Factor-3/genetics , Gene Expression Regulation, Neoplastic , Mitosis/genetics , Polymorphism, Single Nucleotide , Ribonucleoproteins, Small Nuclear/genetics , Breast Neoplasms/epidemiology , Breast Neoplasms/pathology , Case-Control Studies , Female , Gene Frequency , Genetic Predisposition to Disease , Haplotypes , Humans , Linkage Disequilibrium , Logistic Models , Midwestern United States/epidemiology , Neoplasm Invasiveness , Odds Ratio , Pedigree , Phenotype , Risk Assessment , Risk Factors
9.
Br J Cancer ; 101(12): 2048-54, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19920816

ABSTRACT

BACKGROUND: In this study we aimed to evaluate the role of a SNP in intron 1 of the ERCC4 gene (rs744154), previously reported to be associated with a reduced risk of breast cancer in the general population, as a breast cancer risk modifier in BRCA1 and BRCA2 mutation carriers. METHODS: We have genotyped rs744154 in 9408 BRCA1 and 5632 BRCA2 mutation carriers from the Consortium of Investigators of Modifiers of BRCA1/2 (CIMBA) and assessed its association with breast cancer risk using a retrospective weighted cohort approach. RESULTS: We found no evidence of association with breast cancer risk for BRCA1 (per-allele HR: 0.98, 95% CI: 0.93-1.04, P = 0.5) or BRCA2 (per-allele HR: 0.97, 95% CI: 0.89-1.06, P = 0.5) mutation carriers. CONCLUSION: This SNP is not a significant modifier of breast cancer risk for mutation carriers, though weak associations cannot be ruled out.


Subject(s)
DNA-Binding Proteins/genetics , Genes, BRCA1 , Genes, BRCA2 , Heterozygote , Mutation , Polymorphism, Single Nucleotide , Cohort Studies , Female , Humans , Retrospective Studies
10.
Br J Cancer ; 101(8): 1461-8, 2009 Oct 20.
Article in English | MEDLINE | ID: mdl-19738611

ABSTRACT

BACKGROUND: Dysregulation of the cell cycle is a hallmark of many cancers including ovarian cancer, a leading cause of gynaecologic cancer mortality worldwide. METHODS: We examined single nucleotide polymorphisms (SNPs) (n=288) from 39 cell cycle regulation genes, including cyclins, cyclin-dependent kinases (CDKs) and CDK inhibitors, in a two-stage study. White, non-Hispanic cases (n=829) and ovarian cancer-free controls (n=941) were genotyped using an Illumina assay. RESULTS: Eleven variants in nine genes (ABL1, CCNB2, CDKN1A, CCND3, E2F2, CDK2, E2F3, CDC2, and CDK7) were associated with risk of ovarian cancer in at least one genetic model. Seven SNPs were then assessed in four additional studies with 1689 cases and 3398 controls. Association between risk of ovarian cancer and ABL1 rs2855192 found in the original population [odds ratio, OR(BB vs AA) 2.81 (1.29-6.09), P=0.01] was also observed in a replication population, and the association remained suggestive in the combined analysis [OR(BB vs AA) 1.59 (1.08-2.34), P=0.02]. No other SNP associations remained suggestive in the replication populations. CONCLUSION: ABL1 has been implicated in multiple processes including cell division, cell adhesion and cellular stress response. These results suggest that characterization of the function of genetic variation in this gene in other ovarian cancer populations is warranted.


Subject(s)
Cell Cycle/genetics , Genetic Predisposition to Disease , Ovarian Neoplasms/genetics , Polymorphism, Single Nucleotide , Cyclin-Dependent Kinases/genetics , Female , Humans , Ovarian Neoplasms/etiology
11.
Am J Epidemiol ; 166(4): 456-64, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17548785

ABSTRACT

Early-life exposures may influence the development of breast cancer. The authors examined the association of childhood and adolescent anthropometric factors, physical activity levels, and diet with adult mammographic breast density, a strong risk factor for breast cancer. Women in the Minnesota Breast Cancer Family Study cohort who had undergone mammograms but had not had breast cancer (n=1,893) formed the sample. Information on adolescent exposures, including relative height, weight, and physical activity at ages 7, 12, and 18 years and diet at age 12-13 years, was self-reported during two follow-up studies (1990-2003). Mammographic percent density was estimated using a computer-assisted thresholding program. Statistical analyses were performed using linear mixed-effects models with two-sided tests. Positive associations with height at ages 7 (p<0.001), 12 (p<0.001), and 18 (p<0.001) years and percent density were evident overall and within menopausal status categories. The minimum difference in percent density between the tallest and shortest girls was 3 percent, with a maximum of 7 percent. Weight at age 12 years (p=0.005) and adiposity at age 12 years (p=0.005) were both inversely associated with adult percent density. Adolescent physical activity and diet were unrelated to percent density. These results suggest that adolescent height, a known risk factor for breast cancer, is also associated with mammographic percent density.


Subject(s)
Body Weights and Measures , Breast/anatomy & histology , Diet , Physical Fitness , Adolescent , Age Factors , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/epidemiology , Breast Neoplasms/pathology , Child , Confounding Factors, Epidemiologic , Female , Humans , Mammography , Middle Aged , Risk Factors
12.
J Med Genet ; 43(1): 74-83, 2006 Jan.
Article in English | MEDLINE | ID: mdl-15923272

ABSTRACT

BACKGROUND: The vast majority of BRCA1 missense sequence variants remain uncharacterized for their possible effect on protein expression and function, and therefore are unclassified in terms of their pathogenicity. BRCA1 plays diverse cellular roles and it is unlikely that any single functional assay will accurately reflect the total cellular implications of missense mutations in this gene. OBJECTIVE: To elucidate the effect of two BRCA1 variants, 5236G>C (G1706A) and 5242C>A (A1708E) on BRCA1 function, and to survey the relative usefulness of several assays to direct the characterisation of other unclassified variants in BRCA genes. METHODS AND RESULTS: Data from a range of bioinformatic, genetic, and histopathological analyses, and in vitro functional assays indicated that the 1708E variant was associated with the disruption of different cellular functions of BRCA1. In transient transfection experiments in T47D and 293T cells, the 1708E product was mislocalised to the cytoplasm and induced centrosome amplification in 293T cells. The 1708E variant also failed to transactivate transcription of reporter constructs in mammalian transcriptional transactivation assays. In contrast, the 1706A variant displayed a phenotype comparable to wildtype BRCA1 in these assays. Consistent with functional data, tumours from 1708E carriers showed typical BRCA1 pathology, while tumour material from 1706A carriers displayed few histopathological features associated with BRCA1 related tumours. CONCLUSIONS: A comprehensive range of genetic, bioinformatic, and functional analyses have been combined for the characterisation of BRCA1 unclassified sequence variants. Consistent with the functional analyses, the combined odds of causality calculated for the 1706A variant after multifactorial likelihood analysis (1:142) indicates a definitive classification of this variant as "benign". In contrast, functional assays of the 1708E variant indicate that it is pathogenic, possibly through subcellular mislocalisation. However, the combined odds of 262:1 in favour of causality of this variant does not meet the minimal ratio of 1000:1 for classification as pathogenic, and A1708E remains formally designated as unclassified. Our findings highlight the importance of comprehensive genetic information, together with detailed functional analysis for the definitive categorisation of unclassified sequence variants. This combination of analyses may have direct application to the characterisation of other unclassified variants in BRCA1 and BRCA2.


Subject(s)
BRCA1 Protein/chemistry , BRCA1 Protein/genetics , Mutation, Missense/genetics , Adult , Aged , Australia , Breast Neoplasms/pathology , Centrosome/metabolism , Female , Genes, Reporter/genetics , Humans , Loss of Heterozygosity/genetics , Middle Aged , Models, Molecular , Protein Processing, Post-Translational , Protein Transport , RNA Splicing/genetics , RNA Stability/genetics , Transcription, Genetic , Transcriptional Activation/genetics
15.
J Natl Cancer Inst ; 93(21): 1633-7, 2001 Nov 07.
Article in English | MEDLINE | ID: mdl-11698567

ABSTRACT

BACKGROUND: In women with a family history of breast cancer, bilateral prophylactic mastectomy is associated with a decreased risk of subsequent breast cancer of approximately 90%. We examined the association between bilateral prophylactic mastectomy and breast cancer risk in women at high risk for breast cancer who also had mutations in BRCA1 and BRCA2 genes. METHODS: We obtained blood samples from 176 of the 214 high-risk women who participated in our previous retrospective cohort study of bilateral prophylactic mastectomy. We used conformation-sensitive gel electrophoresis and direct sequence analysis of the blood specimens to identify women with mutations in BRCA1 and BRCA2. The carriers' probabilities of developing breast cancer were estimated from two different penetrance models. RESULTS: We identified 26 women with an alteration in BRCA1 or BRCA2. Eighteen of the mutations were considered to be deleterious and eight to be of uncertain clinical significance. None of the 26 women has developed breast cancer after a median of 13.4 years of follow-up (range, 5.8-28.5 years). Three of the 214 women are known to have developed a breast cancer after prophylactic mastectomy. For two of these women, BRCA1 and BRCA2 screening was negative, and no blood specimen was available for the third. Estimations of the effectiveness of prophylactic mastectomy were performed, considering this woman as both a mutation carrier and a noncarrier. These calculations predicted that six to nine breast cancers should have developed among the mutation carriers, which translates into a risk reduction, after bilateral prophylactic mastectomy, of 89.5%-100% (95% confidence interval = 41.4% to 100%). CONCLUSIONS: Prophylactic mastectomy is associated with a substantial reduction in the incidence of subsequent breast cancer not only in women identified as being at high risk on the basis of a family history of breast cancer but also in known BRCA1 or BRCA2 mutation carriers.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/prevention & control , Genes, BRCA1 , Heterozygote , Mastectomy , Mutation , Breast Neoplasms/epidemiology , Female , Genes, BRCA2 , Humans , Incidence
16.
Cancer Res ; 61(15): 5895-904, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11479231

ABSTRACT

In the United States, ovarian cancer is the fourth most common cause of cancer-related deaths among women. The most important prognostic factor for this cancer is tumor stage, or extent of disease at diagnosis. Although women with low-stage tumors have a relatively good prognosis, most women diagnosed with late-stage disease eventually succumb to their cancer. In an attempt to understand early events in ovarian carcinogenesis, and to explore steps in its progression, we have applied multiple molecular genetic techniques to the analysis of 21 early-stage (stage I/II) and 17 advanced-stage (stage III/IV) ovarian tumors. These techniques included expression profiling with cDNA microarrays containing approximately 18,000 expressed sequences, and comparative genomic hybridization to address the chromosomal locations of copy number gains as well as losses. Results from the analysis indicate that early-stage ovarian cancers exhibit profound alterations in gene expression, many of which are similar to those identified in late-stage tumors. However, differences observed at the genomic level suggest differences between the early- and late-stage tumors and provide support for a progression model for ovarian cancer development.


Subject(s)
Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Disease Progression , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genome, Human , Humans , Neoplasm Staging , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
17.
Cancer Res ; 61(13): 4951-5, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11431322

ABSTRACT

A novel region of amplification in breast tumors has recently been identified on chromosome 17q22-23. In an effort to identify the oncogenes in the region that are targeted by the amplification process, we determined the structure of the amplicon in breast cancer cell lines and tumors. Physical and transcription maps of the approximately 3.5-Mb region were established and used as the basis for copy number analysis within the region by Southern blot and fluorescence in situ hybridization. Seven specific and independent amplification maxima were identified in breast cancer cell lines and breast tumors. We present correlative amplification and overexpression studies for the FLJ21316 and Hs.6649 genes suggesting a role for these candidates as amplification-dependent oncogenes.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , Chromosomes, Human, Pair 17/genetics , Gene Amplification , Amino Acid Sequence , Gene Expression Regulation, Neoplastic , Humans , Molecular Sequence Data , Oncogenes , Physical Chromosome Mapping , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
18.
Cancer Epidemiol Biomarkers Prev ; 10(4): 327-32, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11319172

ABSTRACT

Most epidemiological studies of cigarette smoking and breast cancer have failed to demonstrate a strong association. Only one study has been performed on women at high genetic risk, and smoking was reported to be a protective factor. To further explore this observation, we examined the association of cigarette smoking with the risk of breast cancer in a historical cohort study of high-risk breast cancer families. A total of 426 families ascertained through a consecutive series of breast cancer patients (probands) between 1944 and 1952 were followed through 1996. Occurrence of breast cancer and detailed smoking histories for sisters, daughters, granddaughters, nieces, and marry-ins were obtained through telephone interviews between 1991 and 1996. Cox proportional hazards regression, accounting for age, birth cohort, and other risk factors, was used to calculate relative risks and 95% confidence intervals (CIs) of breast cancer. All of the models were constructed within strata defined by relationship to the index case (proband), with nonsmokers designated as the referent group. Of the 426 families in the cohort, 132 had at least three incident breast and/or ovarian cancers in the biological relatives at the end of the follow-up period. Among sisters and daughters in these 132 high-risk families, those who ever smoked were at 2.4-fold increased risk of breast cancer (95% CI, 1.2-5.1) relative to never-smokers. No association between breast cancer and smoking was observed among nieces and granddaughters of probands or among marry-ins. When the analysis was restricted to 35 families at highest genetic risk (each containing five breast and/or ovarian cancers), smoking became an even stronger risk factor. Among sisters and daughters, ever-smokers were at 5.8-fold greater risk than nonsmokers (95% CI, 1.4-23.9). Among nieces and granddaughters, the risk of breast cancer associated with smoking was increased 60% (95% CI, 0.8-3.2). These results suggest that smoking may increase risk for breast cancer in families with multiple cases of breast or ovarian cancer, especially those with the strongest apparent familial predisposition.


Subject(s)
Breast Neoplasms/etiology , Genetic Predisposition to Disease , Ovarian Neoplasms/genetics , Smoking/adverse effects , Adolescent , Adult , Aged , Aged, 80 and over , Breast Neoplasms/genetics , Cohort Studies , Female , Humans , Middle Aged , Ovarian Neoplasms/etiology , Pedigree , Risk Factors
19.
Am J Clin Pathol ; 115(2): 213-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11211609

ABSTRACT

PS6K, a putative oncogene mapped to chromosome 17q23, encodes a serine/threonine kinase, which phosphorylates ribosomal subunit 6 and is part of the insulin receptor signal transduction pathway involved in the regulation of messenger RNA translation, protein synthesis, cell cycle progression, and cell size. Comparative genomic hybridization studies have detected 17q23 amplifications in a subset of meningiomas, particularly those with aggressive histologic features. PS6K amplifications have been reported in breast cancer, another hormonally driven neoplasm. We assessed PS6K dosage in 94 archival paraffin-embedded meningiomas using dual-color fluorescence in situ hybridization. We found high-level PS6K amplifications in 3 of 22 anaplastic grade III meningiomas. Amplification was confirmed by differential polymerase chain reaction in 1 of these cases. In contrast, no amplifications were identified in 37 benign (grade I) and 35 atypical (grade II) meningiomas. To our knowledge, this represents the first report of gene amplification in primary human meningiomas. Given its exclusive association with anaplastic meningiomas, PS6K amplification likely occurs during the malignant progression of a small subset of anaplastic tumors. Further studies are needed to map the 17q23 amplicon to determine whether additional genes in this region are amplified in high-grade meningiomas.


Subject(s)
Gene Amplification , Meningeal Neoplasms/pathology , Meningioma/pathology , Plant Proteins , Protein Serine-Threonine Kinases/genetics , DNA Primers/chemistry , DNA, Neoplasm/analysis , Humans , In Situ Hybridization, Fluorescence , Meningeal Neoplasms/classification , Meningeal Neoplasms/genetics , Meningioma/classification , Meningioma/genetics , Neoplasm Staging , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/metabolism
20.
Cancer Res ; 60(19): 5371-5, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11034073

ABSTRACT

Amplification of the 17q23 region occurs frequently in breast tumors. To characterize the structure of 17q23 amplicons and to identify oncogene targets associated with this alteration, we performed a copy number analysis of 87 17q23 localized expressed sequence tags in seven breast cancer cell lines. Three major regions of amplification were detected in the MCF7 and BT474 cell lines. Amplification of at least one of four known genes (PAT1, PS6K, RAD51C, and SIGMA1B) was detected in the cell lines and in 28% of 94 breast tumors. In most cases, these four genes were overexpressed when amplified, but there was a particularly good association between amplification of the SIGMA1B gene and elevated expression in tumors, which suggested a possible role for this gene in tumor progression. Our data show that this region contains at least four independent targets of amplification, which suggests that there is considerable variability in the structure of the 17q23 amplicon.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , Chromosomes, Human, Pair 17/genetics , Saccharomyces cerevisiae Proteins , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Gene Amplification , Gene Expression , Humans , RNA-Binding Proteins , Rad51 Recombinase , Ribosomal Protein S6 Kinases/biosynthesis , Ribosomal Protein S6 Kinases/genetics , Transcription Factor AP-1/biosynthesis , Transcription Factor AP-1/genetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...