Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Sci Rep ; 7(1): 4713, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28680148

ABSTRACT

Pathogenic BRCA1/2 germline mutations confer high risks of breast and ovarian cancer to women of European ancestry. Characterization of BRCA1/2 mutations in other ethnic groups is also medically important. We comprehensively screened 68 Colombian breast/ovarian cancer families for small-range mutations, 221 families for large-genomic rearrangements, and 1,022 unselected breast cancer cases for Colombian founder mutations in BRCA1/2. The risk of cancer among relatives of mutation carriers and the mutation penetrance were estimated by survival analysis. Identified BRCA2 mutations included 6310delGA and the recurrent 1991del4 mutations. A novel large BRCA2 deletion was found in 0.9% of the screened families. Among unselected breast cancer cases, 3.3% tested positive for BRCA1/3450del4, 2.2% for BRCA1/A1708E, 1.1% for BRCA2/3034del4, and 0.4% for BRCA2/1991del4. Female relatives of carriers of BRCA1/2 founder mutations showed a 5.90 times higher risk of breast cancer, when the woman herself carried a BRCA1 mutation compared to a non-carrier (95% CI 2.01-17.3). The estimated cumulative risk of breast cancer by age 70 years for BRCA1 mutations carriers was 14% (95% CI 5-38) compared to 3% for the general Colombian population (relative risk of breast cancer 4.05). Together with known founder mutations, reported novel variants may ease a cost-effective BRCA1/2 screening in women with Colombian ancestry.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , Breast Neoplasms, Male/genetics , Breast Neoplasms/genetics , Germ-Line Mutation , Ovarian Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Colombia , Female , Founder Effect , Humans , Male , Middle Aged , Pedigree , Penetrance , Prevalence , Survival Analysis
3.
Br J Cancer ; 110(4): 1088-100, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24548884

ABSTRACT

BACKGROUND: Breast cancer is one of the most common malignancies in women. Genome-wide association studies have identified FGFR2 as a breast cancer susceptibility gene. Common variation in other fibroblast growth factor (FGF) receptors might also modify risk. We tested this hypothesis by studying genotyped single-nucleotide polymorphisms (SNPs) and imputed SNPs in FGFR1, FGFR3, FGFR4 and FGFRL1 in the Breast Cancer Association Consortium. METHODS: Data were combined from 49 studies, including 53 835 cases and 50 156 controls, of which 89 050 (46 450 cases and 42 600 controls) were of European ancestry, 12 893 (6269 cases and 6624 controls) of Asian and 2048 (1116 cases and 932 controls) of African ancestry. Associations with risk of breast cancer, overall and by disease sub-type, were assessed using unconditional logistic regression. RESULTS: Little evidence of association with breast cancer risk was observed for SNPs in the FGF receptor genes. The strongest evidence in European women was for rs743682 in FGFR3; the estimated per-allele odds ratio was 1.05 (95% confidence interval=1.02-1.09, P=0.0020), which is substantially lower than that observed for SNPs in FGFR2. CONCLUSION: Our results suggest that common variants in the other FGF receptors are not associated with risk of breast cancer to the degree observed for FGFR2.


Subject(s)
Breast Neoplasms/genetics , Genetic Predisposition to Disease , Receptor, Fibroblast Growth Factor, Type 2/genetics , Case-Control Studies , Female , Genetic Variation , Genome-Wide Association Study , Genotype , Humans , Polymorphism, Single Nucleotide/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 4/genetics , Receptor, Fibroblast Growth Factor, Type 5/genetics
4.
Clin Pharmacol Ther ; 95(2): 216-27, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24060820

ABSTRACT

The International Tamoxifen Pharmacogenomics Consortium was established to address the controversy regarding cytochrome P450 2D6 (CYP2D6) status and clinical outcomes in tamoxifen therapy. We performed a meta-analysis on data from 4,973 tamoxifen-treated patients (12 globally distributed sites). Using strict eligibility requirements (postmenopausal women with estrogen receptor-positive breast cancer, receiving 20 mg/day tamoxifen for 5 years, criterion 1); CYP2D6 poor metabolizer status was associated with poorer invasive disease-free survival (IDFS: hazard ratio = 1.25; 95% confidence interval = 1.06, 1.47; P = 0.009). However, CYP2D6 status was not statistically significant when tamoxifen duration, menopausal status, and annual follow-up were not specified (criterion 2, n = 2,443; P = 0.25) or when no exclusions were applied (criterion 3, n = 4,935; P = 0.38). Although CYP2D6 is a strong predictor of IDFS using strict inclusion criteria, because the results are not robust to inclusion criteria (these were not defined a priori), prospective studies are necessary to fully establish the value of CYP2D6 genotyping in tamoxifen therapy.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Cytochrome P-450 CYP2D6/genetics , Tamoxifen/therapeutic use , Aged , Antineoplastic Agents, Hormonal/pharmacokinetics , Breast Neoplasms/genetics , Female , Genetic Variation/genetics , Genotype , Humans , Menopause , Middle Aged , Pharmacogenetics/methods , Survival Analysis , Tamoxifen/pharmacokinetics , Treatment Outcome
5.
Br J Cancer ; 106(12): 2016-24, 2012 Jun 05.
Article in English | MEDLINE | ID: mdl-22669161

ABSTRACT

BACKGROUND: The variable penetrance of breast cancer in BRCA1/2 mutation carriers suggests that other genetic or environmental factors modify breast cancer risk. Two genes of special interest are prohibitin (PHB) and methylene-tetrahydrofolate reductase (MTHFR), both of which are important either directly or indirectly in maintaining genomic integrity. METHODS: To evaluate the potential role of genetic variants within PHB and MTHFR in breast and ovarian cancer risk, 4102 BRCA1 and 2093 BRCA2 mutation carriers, and 6211 BRCA1 and 2902 BRCA2 carriers from the Consortium of Investigators of Modifiers of BRCA1 and BRCA2 (CIMBA) were genotyped for the PHB 1630 C>T (rs6917) polymorphism and the MTHFR 677 C>T (rs1801133) polymorphism, respectively. RESULTS: There was no evidence of association between the PHB 1630 C>T and MTHFR 677 C>T polymorphisms with either disease for BRCA1 or BRCA2 mutation carriers when breast and ovarian cancer associations were evaluated separately. Analysis that evaluated associations for breast and ovarian cancer simultaneously showed some evidence that BRCA1 mutation carriers who had the rare homozygote genotype (TT) of the PHB 1630 C>T polymorphism were at increased risk of both breast and ovarian cancer (HR 1.50, 95%CI 1.10-2.04 and HR 2.16, 95%CI 1.24-3.76, respectively). However, there was no evidence of association under a multiplicative model for the effect of each minor allele. CONCLUSION: The PHB 1630TT genotype may modify breast and ovarian cancer risks in BRCA1 mutation carriers. This association need to be evaluated in larger series of BRCA1 mutation carriers.


Subject(s)
Breast Neoplasms/genetics , Genes, BRCA1 , Genes, BRCA2 , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Ovarian Neoplasms/genetics , Polymorphism, Genetic , Repressor Proteins/genetics , Female , Genetic Predisposition to Disease , Heterozygote , Humans , Mutation , Prohibitins , Risk
6.
Br J Cancer ; 105(12): 1934-9, 2011 Dec 06.
Article in English | MEDLINE | ID: mdl-22033276

ABSTRACT

BACKGROUND: Somatic mutations in phosphoinositide-3-kinase catalytic subunit alpha (PIK3CA) are frequent in breast tumours and have been associated with oestrogen receptor (ER) expression, human epidermal growth factor receptor-2 overexpression, lymph node metastasis and poor survival. The goal of this study was to evaluate the association between inherited variation in this oncogene and risk of breast cancer. METHODS: A single-nucleotide polymorphism from the PIK3CA locus that was associated with breast cancer in a study of Caucasian breast cancer cases and controls from the Mayo Clinic (MCBCS) was genotyped in 5436 cases and 5280 controls from the Cancer Genetic Markers of Susceptibility (CGEMS) study and in 30 949 cases and 29 788 controls from the Breast Cancer Association Consortium (BCAC). RESULTS: Rs1607237 was significantly associated with a decreased risk of breast cancer in MCBCS, CGEMS and all studies of white Europeans combined (odds ratio (OR)=0.97, 95% confidence interval (CI) 0.95-0.99, P=4.6 × 10(-3)), but did not reach significance in the BCAC replication study alone (OR=0.98, 95% CI 0.96-1.01, P=0.139). CONCLUSION: Common germline variation in PIK3CA does not have a strong influence on the risk of breast cancer.


Subject(s)
Breast Neoplasms/enzymology , Genetic Predisposition to Disease , Genetic Variation , Phosphatidylinositol 3-Kinases/genetics , Breast Neoplasms/genetics , Case-Control Studies , Class I Phosphatidylinositol 3-Kinases , Female , Humans
7.
Br J Cancer ; 101(8): 1456-60, 2009 Oct 20.
Article in English | MEDLINE | ID: mdl-19707196

ABSTRACT

BACKGROUND: The TP53 pathway, in which TP53 and its negative regulator MDM2 are the central elements, has an important role in carcinogenesis, particularly in BRCA1- and BRCA2-mediated carcinogenesis. A single nucleotide polymorphism (SNP) in the promoter region of MDM2 (309T>G, rs2279744) and a coding SNP of TP53 (Arg72Pro, rs1042522) have been shown to be of functional significance. METHODS: To investigate whether these SNPs modify breast cancer risk for BRCA1 and BRCA2 mutation carriers, we pooled genotype data on the TP53 Arg72Pro SNP in 7011 mutation carriers and on the MDM2 309T>G SNP in 2222 mutation carriers from the Consortium of Investigators of Modifiers of BRCA1/2 (CIMBA). Data were analysed using a Cox proportional hazards model within a retrospective likelihood framework. RESULTS: No association was found between these SNPs and breast cancer risk for BRCA1 (TP53: per-allele hazard ratio (HR)=1.01, 95% confidence interval (CI): 0.93-1.10, P(trend)=0.77; MDM2: HR=0.96, 95%CI: 0.84-1.09, P(trend)=0.54) or for BRCA2 mutation carriers (TP53: HR=0.99, 95%CI: 0.87-1.12, P(trend)=0.83; MDM2: HR=0.98, 95%CI: 0.80-1.21, P(trend)=0.88). We also evaluated the potential combined effects of both SNPs on breast cancer risk, however, none of their combined genotypes showed any evidence of association. CONCLUSION: There was no evidence that TP53 Arg72Pro or MDM2 309T>G, either singly or in combination, influence breast cancer risk in BRCA1 or BRCA2 mutation carriers.


Subject(s)
Breast Neoplasms/genetics , Genes, BRCA1 , Genes, BRCA2 , Genes, p53 , Genetic Predisposition to Disease , Mutation , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-mdm2/genetics , Breast Neoplasms/etiology , Female , Heterozygote , Humans , Risk Factors
8.
BJOG ; 116(8): 1053-61, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19438492

ABSTRACT

OBJECTIVE: There is evidence that estrogens and some of their metabolites are involved in endometrial cancer pathogenesis. As estrogens mediate their effects via the estrogen receptors, ESR1 and ESR2, the objective of this investigation was to determine whether six single nucleotide polymorphisms (SNPs) in these two genes were over-represented in a population of endometrial cancer patients compared with a healthy matched control population, thereby associating differences in these genes with endometrial cancer. DESIGN: The study is a case-control investigation large enough to detect a two-fold increased risk, assuming a dominant genetic model, with P = 0.05 and 80% power. SETTING: The study and control populations were all from the Hunter-New England region of New South Wales, Australia collected between the years 1992 and 2005. POPULATION: The study consisted of 191 endometrial cancer patients and 291 healthy controls matched for gender and age. METHODS: Two SNPs in ESR1 and four SNPs in ESR2 were genotyped using PCR-based restriction fragment length polymorphism analysis and real-time PCR. Odds ratios were calculated using unconditional logistic regression and SIMHAP was used for haplotype analysis, adjusting for potential endometrial cancer risk factors. Kaplan-Meier survival analysis, Cox regression and t tests were used to examine the patient's age of diagnosis of endometrial cancer and genotype. MAIN OUTCOME MEASURES: Over-representation of ESR1 and ESR2 polymorphisms in the endometrial cancer population compared with the control population indicates an involvement in the development and/or progression of disease. RESULTS: Two ESR1 (rs2234693 and rs9340799) and two ESR2 (rs1255998 and rs944050) polymorphisms were associated with an increased risk of endometrial cancer. Following adjustment for risk factors, the association with the ESR1 and ESR2 polymorphisms (rs2234693, rs1255998 and rs944050) remained highly significant. Haplotype analysis revealed that carriers of the ESR1 haplotype (variant alleles; rs2234693 and rs9340799) and the ESR2 haplotype (variant allele; rs1255998 and wild-type alleles; rs944050, rs4986938 and rs1256049) were at an increased risk (OR 1.862, P = 0.013 and OR 1.918, P = 0.046 respectively). This risk was even greater in women carrying both risk haplotypes (OR 5.041, P = 0.007). CONCLUSIONS: Our data suggest that the ESR1 (rs2234693 and rs9340799) and the ESR2 (rs1255998 and rs944050) polymorphisms may be associated with an increased risk of developing endometrial cancer.


Subject(s)
Endometrial Neoplasms/genetics , Polymorphism, Single Nucleotide/genetics , Receptors, Estrogen/genetics , Case-Control Studies , Female , Gene Frequency , Genotype , Haplotypes/genetics , Humans , Linkage Disequilibrium , Risk Factors
9.
Br J Cancer ; 99(6): 974-7, 2008 Sep 16.
Article in English | MEDLINE | ID: mdl-18781154

ABSTRACT

The close functional relationship between p53 and the breast cancer susceptibility genes BRCA1 and BRCA2 has promoted the investigation of various polymorphisms in the p53 gene as possible risk modifiers in BRCA1/2 mutation carriers. Specifically, two polymorphisms in p53, c.97-147ins16bp and p.Arg72Pro have been analysed as putative breast cancer susceptibility variants, and it has been recently reported that a p53 haplotype combining the absence of the 16-bp insertion and the presence of proline at codon 72 (No Ins-72Pro) was associated with an earlier age at the onset of the first primary tumour in BRCA2 mutation carriers in the Spanish population. In this study, we have evaluated this association in a series of 2932 BRCA1/2 mutation carriers from the Consortium of Investigators of Modifiers of BRCA1 and BRCA2.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , Breast Neoplasms/genetics , Germ-Line Mutation/genetics , Polymorphism, Genetic/genetics , Tumor Suppressor Protein p53/genetics , Adult , Breast Neoplasms/pathology , Female , Genetic Predisposition to Disease , Genotype , Haplotypes , Heterozygote , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Risk Factors
11.
Cancer Genet Cytogenet ; 129(2): 145-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11566345

ABSTRACT

Chromosome arm 17p is frequently altered in a variety of human cancers, especially in breast cancer, and allelic imbalances (AIs) in the region 17p13.1 do not always coincide with mutations in the TP53 gene. A second interval that frequently shows AIs at 17p is the chromosomal band 17p13.3. This region is suspected to harbor another tumor suppressor gene. In order to get more information concerning the pattern of AIs in 17p13.3, we performed analysis of AI of 49 breast carcinomas at 6 polymorphic loci in 17p13.3. Eighty-six percent of the tumors present AI at least at one marker in 17p13.3. Among all loci tested, the highest percentage of Al was observed at loci D17S5 (77%) and D17S1528 (72%). According to these results, a minimal region of deletion could be determined between the markers D17S28 and D17S5. Fine mapping of this region revealed that the size of the deleted region is about 100-150 kb. Furthermore, a subset of the patients shows two other areas with AI close to the markers D17S1574/D17S513 and D17S849, respectively.


Subject(s)
Allelic Imbalance/genetics , Breast Neoplasms/genetics , Carcinoma/genetics , Chromosomes, Human, Pair 17/genetics , Physical Chromosome Mapping , Female , Genes, Tumor Suppressor , Genetic Markers , Humans , Polymorphism, Genetic , Sequence Deletion/genetics
12.
Clin Lab ; 46(9-10): 447-61, 2000.
Article in English | MEDLINE | ID: mdl-11034530

ABSTRACT

About one in eight to ten women living in Western countries will develop breast cancer during her lifetime and between 5-10% of these cases result from an inherited susceptibility to the disease. Within the past few years, a number of genes associated with a high risk of breast cancer have been identified, including BRCA1, BRCA2, TP53, PTEN, MLH1, MSH2, and STK11. The identification of these genes, together with the rapid advances in molecular genetic analyses, should improve the diagnosis and therapy of breast cancer. This article reviews the genetic basis of hereditary breast cancer, in particular the contribution of BRCA1 and BRCA2 and discusses the clinical application of this new molecular knowledge with regard to molecular testing, surveillance and prevention in women with a hereditary predisposition to breast cancer.


Subject(s)
Breast Neoplasms/genetics , BRCA2 Protein , Breast Neoplasms/diagnosis , Family Health , Female , Genes, BRCA1 , Genetic Markers , Genetic Predisposition to Disease , Genetic Testing , Humans , Male , Neoplasm Proteins/genetics , Pedigree , Transcription Factors/genetics
13.
Breast Cancer Res Treat ; 59(2): 185-92, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10817354

ABSTRACT

Reports from different countries have been inconclusive in attempting to relate the BRCA1 mutation status to the survival of breast cancer patients. The purpose of this study was to investigate overall and disease-free survival for German hereditary breast cancer patients. Data on clinical outcome and data on age at diagnosis of breast cancer, histology, tumor size, lymph node status, histological grade, and laterality of 36 breast cancer patients from 12 families with a BRCA1 mutation and from one family with strong evidence for linkage to BRCA1 were compared with those of 49 hereditary breast cancer patients from 23 families that did not harbor a BRCA1 mutation. Overall and disease-free survival was estimated for both groups. BRCA1 mutation carriers had a significantly earlier age of diagnosis than non-carriers (p = 0.0001) and more frequently developed contralateral breast cancer (p = 0.04). Also, BRCA1-associated tumors more frequently were of larger size (p = 0.041) and higher grade of malignancy (p = 0.005) than non-BRCA1-associated tumors. Whereas no difference in overall survival was seen, disease-free survival at 10 years differed significantly with 53.3% for BRCA1 mutation carriers and 76% for non-carriers (p = 0.02). However, after stratification for age and in multivariate analysis for mutation status, age, and bilaterality, it was shown that the worse prognosis for BRCA1 mutation carriers disappeared. Our results suggest that the worse prognosis of BRCA1 mutation carriers in terms of disease-free survival may in large part be due to the age of onset of breast cancer in this population. Thus, BRCA1 mutation status does not appear to be an independent prognostic factor.


Subject(s)
Breast Neoplasms/genetics , Genes, BRCA1/genetics , Genetic Predisposition to Disease , Point Mutation , Adult , Age of Onset , Breast Neoplasms/pathology , DNA Mutational Analysis , Female , Humans , Middle Aged , Neoplasm Staging , Prognosis , Survival Analysis
14.
J Natl Cancer Inst ; 90(15): 1138-45, 1998 Aug 05.
Article in English | MEDLINE | ID: mdl-9701363

ABSTRACT

BACKGROUND: We have previously demonstrated that breast cancers associated with inherited BRCA1 and BRCA2 gene mutations differ from each other in their histopathologic appearances and that each of these types differs from breast cancers in patients unselected for family history (i.e., sporadic cancers). We have now conducted a more detailed examination of cytologic and architectural features of these tumors. METHODS: Specimens of tumor tissue (5-microm-thick sections) were examined independently by two pathologists, who were unaware of the case or control subject status, for the presence of cell mitosis, lymphocytic infiltration, continuous pushing margins, and solid sheets of cancer cells; cell nuclei, cell nucleoli, cell necrosis, and cell borders were also evaluated. The resulting data were combined with previously available information on tumor type and tumor grade and further evaluated by multifactorial analysis. All statistical tests are two-sided. RESULTS: Cancers associated with BRCA1 mutations exhibited higher mitotic counts (P = .001), a greater proportion of the tumor with a continuous pushing margin (P<.0001), and more lymphocytic infiltration (P = .002) than sporadic (i.e., control) cancers. Cancers associated with BRCA2 mutations exhibited a higher score for tubule formation (fewer tubules) (P = .0002), a higher proportion of the tumor perimeter with a continuous pushing margin (P<.0001), and a lower mitotic count (P = .003) than control cancers. CONCLUSIONS: Our study has identified key features of the histologic phenotypes of breast cancers in carriers of mutant BRCA1 and BRCA2 genes. This information may improve the classification of breast cancers in individuals with a family history of the disease and may ultimately aid in the clinical management of patients.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Genes, BRCA1 , Mutation , Neoplasm Proteins/genetics , Transcription Factors/genetics , Adult , Age Factors , Aged , BRCA2 Protein , Female , Humans , Middle Aged , Multivariate Analysis
15.
Cancer Res ; 58(5): 1004-12, 1998 Mar 01.
Article in English | MEDLINE | ID: mdl-9500463

ABSTRACT

The pattern of loss of heterozygosity (LOH) on chromosome 17 in human breast cancer is complicated and shows many different regions of loss. In an attempt to narrow down the relevant regions of LOH on chromosome 17, we have studied the deletion pattern and its association with clinical parameters in 1280 breast carcinoma-venous blood lymphocyte pairs. In total, 42 different chromosome 17 loci were investigated, and between 25 and 625 cases were analyzed at each locus. The frequency of LOH observed on the p arm was much higher than that observed on the q arm. The opposite effect was observed in 52 ovarian cancer cases investigated, with less LOH on 17p than on 17q. Patterns of loss consistent with interstitial and terminal deletions, as well as loss of either the p or q arm or monosomy 17 were observed. To determine whether loss at particular loci may be associated with biological features of breast tumors, clinical data including age of onset, family history of breast cancer, tumor histopathology, tumor size, estrogen receptor (ER) status, and occurrence of lymph node or distant metastases were collected for each case. Overall, large-sized, ER-negative, lymph node-positive ductal tumors showed the highest frequencies of LOH, with ER-negative and ductal tumors showing LOH for markers along the majority of the chromosome. Eight regions of chromosome 17 appear to be associated with human breast cancer, two on 17p and six on 17q. These regions were not necessarily in the areas exhibiting the highest frequencies of LOH but were defined by interstitial and terminal deletions in multiple independent cases. Seven of these regions showed statistically significant differences in LOH associated with clinical parameters. These data strongly suggest that loci on chromosome 17 may determine aspects of tumor presentation and disease behavior in human breast cancer and pinpoint candidate tumor suppressor gene loci.


Subject(s)
Alleles , Breast Neoplasms/genetics , Chromosomes, Human, Pair 17 , Loss of Heterozygosity , Adult , Breast Neoplasms/pathology , Female , Genes, Tumor Suppressor , Genetic Markers , Humans , Middle Aged , Neoplasm Metastasis/genetics
16.
J Med Genet ; 34(11): 884-8, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9391879

ABSTRACT

In this study we investigated 45 German breast/ovarian cancer families for germline mutations in the BRCA1 gene. We identified four germline mutations in three breast cancer families and in one breast-ovarian cancer family. among these were one frameshift mutation, one nonsense mutation, one novel splice site mutation, and one missense mutation. The missense mutation was also found in 2.8% of the general population, suggesting that it is not disease associated. The average age of disease onset in those families harbouring causative mutations was between 32.3 and 37.4 years, whereas the family harbouring the missense mutation had an average age of onset of 51.2 years. These findings show that BRCA1 is implicated in a small fraction of breast/ovarian cancer families suggesting the involvement of another susceptibility gene(s).


Subject(s)
BRCA1 Protein/genetics , Breast Neoplasms/genetics , Germ-Line Mutation , Ovarian Neoplasms/genetics , Adult , Age of Onset , Aged , Breast Neoplasms, Male/genetics , Chromosomes, Human, Pair 17 , Female , Gene Frequency , Germany , Humans , Male , Middle Aged , Pedigree
17.
Genes Chromosomes Cancer ; 18(3): 193-9, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9071572

ABSTRACT

Loss of heterozygosity (LOH) at 7q31 has been claimed to occur in over 80% of all breast cancers and to be of prognostic significance. This would make this genetic alteration the most common event observed in breast cancer to date. Others, however, have been unable to confirm this high incidence. In this multicenter study, we have complied LOH scorings for three polymorphic markers for 7q31-q32 in 683 breast tumors. Although some significant differences between centers existed, no center reported more than 40% LOH, and the average rate was 19%. Disease-free and overall survival of the patients whose tumors carried LOH at 7q31 did not differ significantly from those patients whose tumors showed retention of heterozygosity at 7q31. In a double-blind scoring of a subset of the raw data, an average discordant rate of LOH scoring of 12% was observed. While startling in itself, this was unable to explain the variation among centers, nor the difference with the initially reported high rate of LOH. We conclude that LOH at 7q31 is not important as a genetic alteration in breast cancer as originally suggested, nor a strong determinant of disease outcome.


Subject(s)
Breast Neoplasms/genetics , Chromosome Aberrations , Chromosomes, Human, Pair 7 , Heterozygote , Breast Neoplasms/mortality , DNA, Neoplasm/analysis , Europe , Humans , Microsatellite Repeats , Prognosis , Survival Analysis
18.
Genes Chromosomes Cancer ; 18(2): 126-32, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9115962

ABSTRACT

Women harboring BRCA1 germline mutations carry an 85% lifetime risk of developing breast cancer and a 63% risk of ovarian cancer. In this first systematic study of familial breast and/or ovarian cancer in Germany we investigated 29 families for germline mutations in the BRCA1 gene. We identified mutations in three breast cancer families and in four breast-ovarian cancer families. The mutations include one missense mutation, one frameshift mutation, one splice mutation, and four nonsense mutations cosegregating with breast and/or ovarian susceptibility in five of ten (50%) families showing positive evidence of linkage to chromosome band 17q21 and in two of 19 (11%) families where linkage data was not available. Two apparently unrelated families carried the same nonsense mutation at codon 1835 and three families harbored a C to T transition at nucleotide 49 of the untranslated exon 4. Allelotyping of the markers D17S855, D17S1322, D17S1323, and D17S1327 located within or near BRCA1 revealed that all affected individuals in the two families harboring the mutation at codon 1835 shared at least one allele indicating a founder mutation. With respect to the overall mutation spectrum, no mutations were identified in exon 11 (0/7) in this set of German families. These findings differed significantly from those in British (17/32)(P = 0.012) and Southern Swedish (13/15) (P < 0.001) families. The lack of BRCA1 mutations in exon 11 which represents 61% of the entire coding sequence may provide additional insight into BRCA1 associated breast and ovarian tumor development.


Subject(s)
BRCA1 Protein/genetics , Breast Neoplasms/genetics , Germ-Line Mutation , Ovarian Neoplasms/genetics , Female , Germany , Humans , Pedigree , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational
19.
J Cancer Res Clin Oncol ; 123(5): 272-9, 1997.
Article in English | MEDLINE | ID: mdl-9201250

ABSTRACT

Female carriers of mutations in the BRCA1 gene on chromosome 17q have a very high risk of developing breast and/or ovarian cancer during their lifetime. There is, however, little knowledge of to what extent non-genetic risk factors, such as age at menarche, age at first birth, and body mass index, alter the age at onset of disease. We identified individuals showing a high probability of linkage to BRCA1 and examined the effect of other known risk factors on disease risk. A total of 43 families with at least three breast or ovarian cancer cases, including two affected before 60 years of age, were studied for linkage to the susceptibility locus BRCA1. Blood samples from relevant family members were used to genotype for at least three chromosome 17q polymorphic markers. Information on reproductive history, hormone use and lifestyle factors was collected from female members using a self-administered questionnaire. Diagnoses of breast and ovarian cancer were verified through pathology reports and paraffin blocks were obtained when available. Multipoint LOD (logarithm of the odds) scores were calculated and individuals from 10 families with a posteriori probability for linkage greater than 0.90 were used for further analysis. Forty-six BRCA1 carriers were identified by the disease haplotype; 30 were affected with breast cancer and 5 with ovarian cancer. Proportional- hazards analysis of age at onset of breast cancer yielded increased relative risks of 1.74 for early age at menarche (< 14 years), 1.58 for late age at first birth (> or = 25 years) or nulliparity, and 2.78 for recent year of birth (> or = 1940); however, none of the risk estimates was statistically significant. When both breast and ovarian cancer were considered as disease endpoints, the birth cohort effect was stronger and age at first birth showed no effect. Our data provide some evidence that reproductive risk factors for breast cancer have an effect on age at onset for BRCA1 carriers. However, considering that our analyses were based on limited numbers, these results warrant further clarification.


Subject(s)
Age of Onset , Breast Neoplasms/epidemiology , Breast Neoplasms/genetics , Genes, BRCA1/genetics , Heterozygote , Mutation , Reproduction , Adult , Aged , Disease Susceptibility , Female , Genetic Linkage , Germany/epidemiology , Humans , Middle Aged , Proportional Hazards Models , Risk , Risk Factors , Survival Analysis
20.
J Med Genet ; 33(8): 633-5, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8863152

ABSTRACT

An estimated 5% to 10% of all breast and ovarian cancers are attributed to dominant susceptibility genes. Two such genes, BRCA1 and BRCA2, were recently identified. The involvement of these genes was studied in 43 German breast only and breast-ovarian cancer families. All families contained three or more cases of breast or ovarian cancer, with at least two diagnosed under the age of 60 years. Multipoint linkage analysis gave a maximum lod score of 2.13 at the BRCA1 locus under the assumption of genetic heterogeneity, with an estimated 50% of families being linked. Among the 33 breast only cancer and 10 breast-ovarian cancer families, the estimated proportions of linked families were 35% and 75%, respectively. Sixteen families with at least four cases of female breast cancer diagnosed under the age of 60 years, or male breast cancer diagnosed at any age, were analysed for linkage to BRCA2. Positive lod scores at BRCA2 were obtained in six families.


Subject(s)
Breast Neoplasms, Male/genetics , Breast Neoplasms/genetics , Genetic Heterogeneity , Lod Score , Ovarian Neoplasms/genetics , Adult , Aged , Aged, 80 and over , BRCA2 Protein , Breast Neoplasms/complications , Female , Genes, BRCA1/genetics , Genetic Markers , Germany , Humans , Male , Middle Aged , Neoplasm Proteins/genetics , Ovarian Neoplasms/complications , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...