Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Gynecol Oncol ; 164(2): 245-253, 2022 02.
Article in English | MEDLINE | ID: mdl-34906376

ABSTRACT

OBJECTIVE: In the Phase 3 VELIA trial (NCT02470585), PARP inhibitor (PARPi) veliparib was combined with first-line chemotherapy and continued as maintenance for patients with ovarian carcinoma enrolled regardless of chemotherapy response or biomarker status. Here, we report exploratory analyses of the impact of homologous recombination deficient (HRD) or proficient (HRP) status on progression-free survival (PFS) and objective response rates during chemotherapy. METHODS: Women with Stage III-IV ovarian carcinoma were randomized to veliparib-throughout, veliparib-combination-only, or placebo. Stratification factors included timing of surgery and germline BRCA mutation status. HRD status was dichotomized at genomic instability score 33. During combination therapy, CA-125 levels were measured at baseline and each cycle; radiographic responses were assessed every 9 weeks. RESULTS: Of 1140 patients randomized, 742 had BRCA wild type (BRCAwt) tumors (HRP, n = 373; HRD/BRCAwt, n = 329). PFS hazard ratios between veliparib-throughout versus control were similar in both BRCAwt populations (HRD/BRCAwt: 22.9 vs 19.8 months; hazard ratio 0.76; 95% confidence interval [CI] 0.53-1.09; HRP: 15.0 vs 11.5 months; hazard ratio 0.765; 95% CI 0.56-1.04). By Cycle 3, the proportion with ≥90% CA-125 reduction from baseline was higher in those receiving veliparib (pooled arms) versus control (34% vs 23%; P = 0.0004); particularly in BRCAwt and HRP subgroups. Complete response rates among patients with measurable disease after surgery were 24% with veliparib (pooled arms) and 18% with control. CONCLUSIONS: These results potentially broaden opportunities for PARPi utilization among patients who would not qualify for frontline PARPi maintenance based on other trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzimidazoles/therapeutic use , Carcinoma, Ovarian Epithelial/drug therapy , Hereditary Breast and Ovarian Cancer Syndrome/drug therapy , Ovarian Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Recombinational DNA Repair/genetics , Adult , Aged , Aged, 80 and over , Allelic Imbalance/genetics , CA-125 Antigen/metabolism , Carboplatin/administration & dosage , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Cytoreduction Surgical Procedures , Female , Genes, BRCA1 , Genes, BRCA2 , Genomic Instability/genetics , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Hereditary Breast and Ovarian Cancer Syndrome/metabolism , Hereditary Breast and Ovarian Cancer Syndrome/pathology , Humans , Induction Chemotherapy , Loss of Heterozygosity/genetics , Maintenance Chemotherapy , Middle Aged , Neoplasm Staging , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Paclitaxel/administration & dosage , Progression-Free Survival , Proportional Hazards Models , Young Adult
2.
Int J Mol Sci ; 21(24)2020 Dec 14.
Article in English | MEDLINE | ID: mdl-33327492

ABSTRACT

Hereditary gynecological cancers are caused by several inherited genes. Tumors that arise in the female reproductive system, such as ovaries and the uterus, overlap with hereditary cancers. Several hereditary cancer-related genes are important because they might lead to therapeutic targets. Treatment of hereditary cancers should be updated in line with the advent of various new methods of evaluation. Next-generation sequencing has led to rapid, economical genetic analyses that have prompted a concomitant and significant paradigm shift with respect to hereditary cancers. Molecular tumor profiling is an epochal method for determining therapeutic targets. Clinical treatment strategies are now being designed based on biomarkers based on tumor profiling. Furthermore, the National Comprehensive Cancer Network (NCCN) guidelines significantly changed the genetic testing process in 2020 to initially consider multi-gene panel (MGP) evaluation. Here, we reviewed the molecular features and clinical management of hereditary gynecological malignancies, such as hereditary breast and ovarian cancer (HBOC), and Lynch, Li-Fraumeni, Cowden, and Peutz-Jeghers syndromes. We also reviewed cancer-susceptible genes revealed by MGP tests.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/metabolism , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Hereditary Breast and Ovarian Cancer Syndrome/pathology , Breast Neoplasms/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Female , Genetic Predisposition to Disease/genetics , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Hereditary Breast and Ovarian Cancer Syndrome/metabolism , High-Throughput Nucleotide Sequencing , Humans
3.
J Pathol ; 248(4): 409-420, 2019 08.
Article in English | MEDLINE | ID: mdl-30883759

ABSTRACT

Splicing disruption is a common mechanism of gene inactivation associated with germline variants of susceptibility genes. To study the role of BRCA2 mis-splicing in hereditary breast/ovarian cancer (HBOC), we performed a comprehensive analysis of variants from BRCA2 exons 2-9, as well as the initial characterization of the regulatory mechanisms of such exons. A pSAD-based minigene with exons 2-9 was constructed and validated in MCF-7 cells, producing the expected transcript (1016-nt/V1-BRCA2_exons_2-9-V2). DNA variants from mutational databases were analyzed by NNSplice and Human Splicing Finder softwares. To refine ESE-variant prediction, we mapped the regulatory regions through a functional strategy whereby 26 exonic microdeletions were introduced into the minigene and tested in MCF-7 cells. Thus, we identified nine spliceogenic ESE-rich intervals where ESE-variants may be located. Combining bioinformatics and microdeletion assays, 83 variants were selected and genetically engineered in the minigene. Fifty-three changes impaired splicing: 28 variants disrupted the canonical sites, four created new ones, 10 abrogated enhancers, eight created silencers and three caused a double-effect. Notably, nine spliceogenic-ESE variants were located within ESE-containing intervals. Capillary electrophoresis and sequencing revealed more than 23 aberrant transcripts, where exon skipping was the most common event. Interestingly, variant c.67G>A triggered the usage of a noncanonical GC-donor 4-nt upstream. Thirty-six variants that induced severe anomalies (>60% aberrant transcripts) were analyzed according to the ACMG guidelines. Thus, 28 variants were classified as pathogenic, five as likely pathogenic and three as variants of uncertain significance. Interestingly, 13 VUS were reclassified as pathogenic or likely pathogenic variants. In conclusion, a large fraction of BRCA2 variants (∼64%) provoked splicing anomalies lending further support to the high prevalence of this disease-mechanism. The low accuracy of ESE-prediction algorithms may be circumvented by functional ESE-mapping that represents an optimal strategy to identify spliceogenic ESE-variants. Finally, systematic functional assays by minigenes depict a valuable tool for the initial characterization of splicing anomalies and the clinical interpretation of variants. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
BRCA2 Protein/genetics , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Gene Deletion , Genes, BRCA2 , Hereditary Breast and Ovarian Cancer Syndrome/genetics , RNA Splicing , BRCA2 Protein/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Computational Biology , Exons , Female , Hereditary Breast and Ovarian Cancer Syndrome/metabolism , Hereditary Breast and Ovarian Cancer Syndrome/pathology , Humans
4.
Mol Med Rep ; 18(2): 1531-1537, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29901183

ABSTRACT

In 2015, according to the National Institute of Statistics and Geography (INEGI), malignant breast tumors were the first cause of cancer fatality in women (6,273 fatalities) in Mexico, whereas 2,793 fatalities in women were due to ovarian cancer. A total of 5­10% of breast cancer and 10­15% of ovarian cancer cases are caused by a hereditary breast­ovarian cancer syndrome, with mutations predominantly identified in the BRCA1 and BRCA2 genes. Recently, the Mexican founder mutation BRCA1 ex9­12del was identified (deletion of exons 9­12 with recombination between introns 8­12). This is the most frequently reported mutation in hereditary breast/ovarian cancer in Mexico. Current detection methods include end­point polymerase chain reaction (PCR) and Multiplex Ligation­dependent Probe Amplification (MLPA). In the present study a cheap, sensitive and fast detection method was developed based on quantitative PCR and two TaqMan® probes, one to detect the deletion (recombination region between introns 8 and 12), and the other one a region from exon 11. With this assay, 90 samples were able to be analyzed in 2 h using 2.5 ng of DNA/reaction at a cost of ~2­3 USD. This method is capable of detecting positive samples for DNA deletion and excluding negative ones. Therefore, the method proposed may be a useful high­throughput diagnostic option that could be useful in future association or prevalence studies that use large populations.


Subject(s)
BRCA1 Protein/genetics , Base Sequence , DNA Probes/chemical synthesis , Hereditary Breast and Ovarian Cancer Syndrome/diagnosis , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Real-Time Polymerase Chain Reaction/methods , Sequence Deletion , Adult , BRCA1 Protein/deficiency , Exons , Female , Gene Expression , Genetic Predisposition to Disease , Genetic Testing , Hereditary Breast and Ovarian Cancer Syndrome/metabolism , Hereditary Breast and Ovarian Cancer Syndrome/pathology , Humans , Introns , Mexico , Middle Aged , Mutation Rate , Sensitivity and Specificity
5.
Sci Rep ; 7(1): 17831, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29259228

ABSTRACT

Hereditary breast and ovarian cancer syndrome (HBOC) is partly due to the presence of mutations in the BRCA genes. Triple-negative (TN) breast cancer (BC) shares histological characteristics with germline BRCA1 mutation-associated tumours. We have investigated the metabolic profiles of human breast cancer (BC) cell lines carrying BRCA1 pathogenic mutations by non-targeted liquid chromatography coupled to mass spectrometry technology. Based on our in vitro results, we performed a targeted metabolomic analysis of plasma samples from TN HBOC patients taking into account their BRCA1 genotype. BRCA1 promoter hypermethylation and the BRCAness phenotype of BC cell lines were also studied. The purpose of this study was to determine the metabolic signature of HBOC syndrome and TNBC patients and to evaluate the potential contribution of the metabolites identified to the genetic diagnosis of breast cancer. The present results show the existence of a differential metabolic signature for BC cells based on the BRCA1 functionality. None of the studied BC cell lines presented hypermethylation of the BRCA1 promoter region. We provide evidence of the existence of free methylated nucleotides capable of distinguishing plasma samples from HBOC patients as BRCA1-mutated and BRCA1 non-mutated, suggesting that they might be considered as BRCA1-like biomarkers for TNBC and HBOC syndrome.


Subject(s)
BRCA1 Protein/genetics , Biomarkers, Tumor/blood , Biomarkers, Tumor/metabolism , Metabolome/genetics , Triple Negative Breast Neoplasms/blood , Triple Negative Breast Neoplasms/metabolism , Cell Line, Tumor , DNA Methylation/genetics , Female , Germ-Line Mutation/genetics , Hereditary Breast and Ovarian Cancer Syndrome/blood , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Hereditary Breast and Ovarian Cancer Syndrome/metabolism , Humans , MCF-7 Cells , Metabolomics/methods , Middle Aged , Phenotype , Promoter Regions, Genetic/genetics , Triple Negative Breast Neoplasms/genetics
6.
BMC Cancer ; 17(1): 496, 2017 Jul 24.
Article in English | MEDLINE | ID: mdl-28738860

ABSTRACT

BACKGROUND: BRCA1 and BRCA2 mutations explain approximately one-fifth of the inherited susceptibility in high-risk Finnish hereditary breast and ovarian cancer (HBOC) families. EMSY is located in the breast cancer-associated chromosomal region 11q13. The EMSY gene encodes a BRCA2-interacting protein that has been implicated in DNA damage repair and genomic instability. We analysed the role of germline EMSY variation in breast/ovarian cancer predisposition. The present study describes the first EMSY screening in patients with high familial risk for this disease. METHODS: Index individuals from 71 high-risk, BRCA1/2-negative HBOC families were screened for germline EMSY sequence alterations in protein coding regions and exon-intron boundaries using Sanger sequencing and TaqMan assays. The identified variants were further screened in 36 Finnish HBOC patients and 904 controls. Moreover, one novel intronic deletion was screened in a cohort of 404 breast cancer patients unselected for family history. Haplotype block structure and the association of haplotypes with breast/ovarian cancer were analysed using Haploview. The functionality of the identified variants was predicted using Haploreg, RegulomeDB, Human Splicing Finder, and Pathogenic-or-Not-Pipeline 2. RESULTS: Altogether, 12 germline EMSY variants were observed. Two alterations were located in the coding region, five alterations were intronic, and five alterations were located in the 3'untranslated region (UTR). Variant frequencies did not significantly differ between cases and controls. The novel variant, c.2709 + 122delT, was detected in 1 out of 107 (0.9%) breast cancer patients, and the carrier showed a bilateral form of the disease. The deletion was absent in 897 controls (OR = 25.28; P = 0.1) and in 404 breast cancer patients unselected for family history. No haplotype was identified to increase the risk of breast/ovarian cancer. Functional analyses suggested that variants, particularly in the 3'UTR, were located within regulatory elements. The novel deletion was predicted to affect splicing regulatory elements. CONCLUSIONS: These results suggest that the identified EMSY variants are likely neutral at the population level. However, these variants may contribute to breast/ovarian cancer risk in single families. Additional analyses are warranted for rare novel intronic deletions and the 3'UTR variants predicted to have functional roles.


Subject(s)
Genetic Predisposition to Disease , Germ-Line Mutation , Hereditary Breast and Ovarian Cancer Syndrome/metabolism , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Repressor Proteins/genetics , Adult , DNA Mutational Analysis , Female , Finland , Genetic Association Studies , Genotyping Techniques , Haplotypes , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Humans , Middle Aged
7.
PLoS One ; 12(1): e0169673, 2017.
Article in English | MEDLINE | ID: mdl-28060958

ABSTRACT

BACKGROUND: Risk-reducing salpingo-oophorectomy (RRSO) reduces ovarian cancer risk in BRCA1/2 mutation carriers. Premenopausal RRSO is hypothesized to increase fracture risk more than natural menopause. Elevated bone turnover markers (BTMs) might predict fracture risk. We investigated BTM levels after RRSO and aimed to identify clinical characteristics associated with elevated BTMs. METHODS: Osteocalcin (OC), procollagen type I N-terminal peptide (PINP) and serum C-telopeptide of type I collagen (sCTx) were measured in 210 women ≥ 2 years after RRSO before age 53. BTM Z-scores were calculated using an existing reference cohort of age-matched women. Clinical characteristics were assessed by questionnaire. RESULTS: BTMs after RRSO were higher than age-matched reference values: median Z-scores OC 0.11, p = 0.003; PINP 0.84, p < 0.001; sCTx 0.53, p < 0.001 (compared to Z = 0). After excluding women with recent fractures or BTM interfering medication, Z-scores increased to 0.34, 1.14 and 0.88, respectively. Z-scores for OC and PINP were inversely correlated to age at RRSO. No correlation was found with fracture incidence or history of breast cancer. CONCLUSIONS: Five years after RRSO, BTMs were higher than age-matched reference values. Since elevated BTMs might predict higher fracture risk, prospective studies are required to evaluate the clinical implications of this finding.


Subject(s)
Bone and Bones/metabolism , Hereditary Breast and Ovarian Cancer Syndrome/etiology , Hereditary Breast and Ovarian Cancer Syndrome/metabolism , Ovariectomy , Salpingectomy , Adult , Aged , Biomarkers , Bone Resorption/metabolism , Collagen Type I/metabolism , Female , Hereditary Breast and Ovarian Cancer Syndrome/epidemiology , Humans , Middle Aged , Osteocalcin/metabolism , Peptide Fragments/metabolism , Peptides/metabolism , Procollagen/metabolism , Risk Factors , Risk Reduction Behavior
8.
EBioMedicine ; 2(10): 1331-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26629528

ABSTRACT

Breast cancer development in BRCA1/2 mutation carriers is a net consequence of cell-autonomous and cell nonautonomous factors which may serve as excellent targets for cancer prevention. In light of our previous data we sought to investigate the consequences of the BRCA-mutation carrier state on RANKL/osteoprotegerin (OPG) signalling. We analysed serum levels of RANKL, OPG, RANKL/OPG complex, oestradiol (E2), and progesterone (P) during menstrual cycle progression in 391 BRCA1/2-mutation carriers and 782 noncarriers. These studies were complemented by analyses of RANKL and OPG in the serum and mammary tissues of female cynomolgus macaques (n = 88) and serum RANKL and OPG in postmenopausal women (n = 150). BRCA-mutation carriers had lower mean values of free serum OPG in particular in BRCA1-mutation carriers (p = 0.018) compared with controls. Among BRCA1/2 mutation carriers, lower OPG levels were associated with germline mutation locations known to confer an increased breast cancer risk (p = 0.003). P is associated with low OPG levels in serum and tissue, particularly in BRCA-mutation carriers (rho = - 0.216; p = 0.002). Serum OPG levels were inversely correlated (rho = - 0.545, p < 0.001) with mammary epithelial proliferation measured by Ki67 expression and increased (p = 0.01) in postmenopause. The P-RANKL/OPG system is dysregulated in BRCA-mutation carriers. These and previously published data provide a strong rationale for further investigation of antiprogestogens or an anti-RANKL antibody such as denosumab for breast cancer prevention.


Subject(s)
Genes, BRCA1 , Genes, BRCA2 , Heterozygote , Mutation , Osteoprotegerin/metabolism , RANK Ligand/metabolism , Animals , Case-Control Studies , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Female , Gene Expression Regulation, Neoplastic , Germ-Line Mutation , Hereditary Breast and Ovarian Cancer Syndrome/blood , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Hereditary Breast and Ovarian Cancer Syndrome/metabolism , Hormones/blood , Humans , Macaca fascicularis , Osteoprotegerin/blood , Patient Outcome Assessment , Postmenopause , RANK Ligand/blood , Risk Factors
9.
Genetics ; 200(2): 413-22, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25823446

ABSTRACT

Interpreting variants of uncertain significance (VUS) is a central challenge in medical genetics. One approach is to experimentally measure the functional consequences of VUS, but to date this approach has been post hoc and low throughput. Here we use massively parallel assays to measure the effects of nearly 2000 missense substitutions in the RING domain of BRCA1 on its E3 ubiquitin ligase activity and its binding to the BARD1 RING domain. From the resulting scores, we generate a model to predict the capacities of full-length BRCA1 variants to support homology-directed DNA repair, the essential role of BRCA1 in tumor suppression, and show that it outperforms widely used biological-effect prediction algorithms. We envision that massively parallel functional assays may facilitate the prospective interpretation of variants observed in clinical sequencing.


Subject(s)
BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Genetic Variation , High-Throughput Nucleotide Sequencing , Protein Interaction Domains and Motifs , BRCA1 Protein/chemistry , DNA Repair , Genomics/methods , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Hereditary Breast and Ovarian Cancer Syndrome/metabolism , Humans , Mutation , Protein Interaction Domains and Motifs/genetics , Protein Multimerization , Ubiquitin-Protein Ligases
10.
BMC Cancer ; 13: 484, 2013 Oct 19.
Article in English | MEDLINE | ID: mdl-24139550

ABSTRACT

BACKGROUND: Most currently known breast cancer predisposition genes play a role in DNA repair by homologous recombination. Recent studies conducted on RAD51 paralogs, involved in the same DNA repair pathway, have identified rare germline mutations conferring breast and/or ovarian cancer predisposition in the RAD51C, RAD51D and XRCC2 genes. The present study analysed the five RAD51 paralogs (RAD51B, RAD51C, RAD51D, XRCC2, XRCC3) to estimate their contribution to breast and ovarian cancer predisposition. METHODS: The study was conducted on 142 unrelated patients with breast and/or ovarian cancer either with early onset or with a breast/ovarian cancer family history. Patients were referred to a French family cancer clinic and had been previously tested negative for a BRCA1/2 mutation. Coding sequences of the five genes were analysed by EMMA (Enhanced Mismatch Mutation Analysis). Detected variants were characterized by Sanger sequencing analysis. RESULTS: Three splicing mutations and two likely deleterious missense variants were identified: RAD51B c.452 + 3A > G, RAD51C c.706-2A > G, RAD51C c.1026 + 5_1026 + 7del, RAD51B c.475C > T/p.Arg159Cys and XRCC3 c.448C > T/p.Arg150Cys. No RAD51D and XRCC2 gene mutations were detected. These mutations and variants were detected in families with both breast and ovarian cancers, except for the RAD51B c.475C > T/p.Arg159Cys variant that occurred in a family with 3 breast cancer cases. CONCLUSIONS: This study identified the first RAD51B mutation in a breast and ovarian cancer family and is the first report of XRCC3 mutation analysis in breast and ovarian cancer. It confirms that RAD51 paralog mutations confer breast and ovarian cancer predisposition and are rare events. In view of the low frequency of RAD51 paralog mutations, international collaboration of family cancer clinics will be required to more accurately estimate their penetrance and establish clinical guidelines in carrier individuals.


Subject(s)
Breast Neoplasms/genetics , DNA-Binding Proteins/genetics , Genetic Predisposition to Disease , Germ-Line Mutation , Breast Neoplasms/metabolism , DNA Mutational Analysis , DNA-Binding Proteins/metabolism , Exons , Female , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Hereditary Breast and Ovarian Cancer Syndrome/metabolism , Humans , Immunohistochemistry , Pedigree , RNA Splicing
11.
Arch Gynecol Obstet ; 288(4): 839-44, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23553196

ABSTRACT

INTRODUCTION: High mortality rate, absence of reliable methods for early diagnosis and poor prognosis of advanced ovarian cancer prompted to investigate the role of prophylactic oophorectomy in BRCA1 mutation carriers as well as evaluate the expression of BRCA1, p53, Nm23, and KAI1 proteins in ovarian tissue from these patients. MATERIALS AND METHODS: Ovaries from BRCA1 mutation carriers underwent prophylactic adnexectomy and control group of patients were operated from other than cancer reasons. The expression of selected proteins was studied using immunohistochemical staining. The intensity of immunostaining and the number of tumor cells showing the reaction for selected proteins were analyzed. RESULTS: We have analyzed ovarian tissues from 18 BRCA1 mutation carriers and 11 women included in control group. Positive expression of BRCA1 protein was presented in 83.3 % cases in BRCA1 mutation carriers and in 72.7 % in the control group (p > 0.05). Positive expression of p53 protein was observed, respectively, in 27.8 vs. 36.4 % (p > 0.05), Nm23 protein 77.7 vs. 90.9 % (p > 0.05), and KAI1 in 72.2 vs. 72.7 % (p > 0.05). Mean percent of tumor cells that showed the reaction for selected proteins as well as the intensity of immunostaining for all analyzed proteins seems to be lower in BRCA1 mutation carriers. CONCLUSIONS: However, any significant differences between study group and control group have not been found; there were similar trends showing reduced expression of studied proteins in BRCA1 mutation carriers.


Subject(s)
Biomarkers, Tumor/metabolism , Hereditary Breast and Ovarian Cancer Syndrome/prevention & control , Ovariectomy , Ovary/metabolism , Salpingectomy , Adult , BRCA1 Protein/metabolism , Case-Control Studies , Female , Genes, BRCA1 , Genetic Predisposition to Disease , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Hereditary Breast and Ovarian Cancer Syndrome/metabolism , Humans , Immunohistochemistry , Kangai-1 Protein/metabolism , Middle Aged , Mutation , NM23 Nucleoside Diphosphate Kinases/metabolism , Ovary/surgery , Tumor Suppressor Protein p53/metabolism
12.
Psychosomatics ; 53(6): 582-90, 2012.
Article in English | MEDLINE | ID: mdl-23157996

ABSTRACT

OBJECTIVE: Cancer risk-related stressors are prominent among BRCA mutation carriers. Loss of one's mother to a BRCA-associated cancer is an additional stressor, which may be related to an enhanced inflammatory response. This study examined the effect of mother's vital status on psychological factors and stress-associated biomarkers among BRCA mutation carriers. The role of bereavement on biopsychological variables was also examined. METHODS: BRCA-carriers with known maternal transmission enrolled in the Gilda Radner Hereditary Cancer Program were invited to participate. Focus group composition was predetermined based on participants' personal cancer history and mother's vital status. Prior to the focus group, participants completed a Quality of Life (QOL) survey and collected a first morning saliva sample. Inflammatory biomarkers were analyzed from proximal archived serum. One day post focus group, a process survey, and morning saliva were collected. RESULTS: QOL was significantly lower for those whose mothers are deceased (n = 17) compared to those whose mothers are alive (n = 15) (P = 0.003) after adjusting for age, personal cancer history and prophylactic surgery. Similarly, those whose mothers are deceased reported significantly more perceived stress (P = 0.015), more intrusive thoughts related to cancer risk (P = 0.049), and more anxiety (P = 0.003). Higher bereavement scores were significantly associated with QOL and psychological measures. Biomarker correlates were consistent with and significantly correlated to the patient-reported psychological outcomes for those whose mothers were deceased. CONCLUSIONS: BRCA mutation carriers with a known maternal transmission whose mother is deceased report higher perceived stress and anxiety, lower QOL, and a stress-associated biomarker profile that is potentially globally immune suppressive.


Subject(s)
Genes, BRCA1 , Genes, BRCA2 , Hereditary Breast and Ovarian Cancer Syndrome/psychology , Maternal Death/psychology , Mutation , Stress, Psychological/psychology , Adult , Antibiotic Prophylaxis , Bereavement , Biomarkers , Female , Focus Groups , Genetic Predisposition to Disease/genetics , Genetic Predisposition to Disease/psychology , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Hereditary Breast and Ovarian Cancer Syndrome/metabolism , Heterozygote , Humans , Hydrocortisone/metabolism , Linear Models , Pilot Projects , Psychometrics , Quality of Life , Saliva/chemistry , Stress, Psychological/metabolism , Surveys and Questionnaires
13.
Breast Cancer Res Treat ; 134(1): 353-62, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22434525

ABSTRACT

In spite the close association of the triple-negative breast cancer immunophenotype with hereditary breast cancers and the BRCA1 pathway, there is a lack of population studies that determine the frequency of BRCA1 mutations among triple-negative breast cancer patients. To address this, we have screened a large sample of 403 women diagnosed with triple-negative invasive breast cancer, independently of their age or family history, for germline BRCA1 mutations. Median age at diagnosis was 50 years (range 20-83). The overall prevalence of triple-negative cases among the initial patient group with invasive breast cancer was 8%. BRCA1 was screened by direct DNA sequencing in all patients, including all exons where a mutation was previously found in the Greek population (exons 5, 11, 12, 16, 20, 21, 22, 23, 24-77% of the BRCA1 coding region), including diagnostic PCRs to detect the three Greek founder large genomic rearrangements. Sixty-five deleterious BRCA1 mutations were identified among the 403 triple-negative breast cancer patients (16%). Median age of onset for mutation carriers was 39 years. Among a total of 106 women with early-onset triple-negative breast cancer (<40 years), 38 (36%) had a BRCA1 mutation, while 27% of women with triple-negative breast cancer diagnosed before 50 years (56/208) had a BRCA1 mutation. A mutation was found in 48% (50/105) of the triple-negative breast cancer patients with family history of breast or ovarian cancer. It is noteworthy, however, that of the 65 carriers, 15 (23%) had no reported family history of related cancers. All but one of the carriers had grade III tumors (98%). These results indicate that women with early-onset triple-negative breast cancer, and ideally all triple-negative breast cancer patients, are candidates for BRCA1 genetic testing even in the absence of a family history of breast or ovarian cancer.


Subject(s)
BRCA1 Protein/genetics , Carcinoma, Ductal, Breast/genetics , Carcinoma, Lobular/genetics , Genetic Testing , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Adult , Aged , Aged, 80 and over , Carcinoma, Ductal, Breast/diagnosis , Carcinoma, Ductal, Breast/epidemiology , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Lobular/diagnosis , Carcinoma, Lobular/epidemiology , Carcinoma, Lobular/metabolism , DNA Mutational Analysis , Female , Hereditary Breast and Ovarian Cancer Syndrome/diagnosis , Hereditary Breast and Ovarian Cancer Syndrome/epidemiology , Hereditary Breast and Ovarian Cancer Syndrome/metabolism , Heterozygote , Humans , Middle Aged , Mutation , Patient Selection , Prevalence , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...