Pathology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 72
  • Item
    Thumbnail Image
    Morphological predictors of BRCA1 germline mutations in young women with breast cancer
    Southey, MC ; Ramus, SJ ; Dowty, JG ; Smith, LD ; Tesoriero, AA ; Wong, EEM ; Dite, GS ; Jenkins, MA ; Byrnes, GB ; Winship, I ; Phillips, K-A ; Giles, GG ; Hopper, JL (NATURE PUBLISHING GROUP, 2011-03-15)
    BACKGROUND: Knowing a young woman with newly diagnosed breast cancer has a germline BRCA1 mutation informs her clinical management and that of her relatives. We sought an optimal strategy for identifying carriers using family history, breast cancer morphology and hormone receptor status data. METHODS: We studied a population-based sample of 452 Australian women with invasive breast cancer diagnosed before age 40 years for whom we conducted extensive germline mutation testing (29 carried a BRCA1 mutation) and a systematic pathology review, and collected three-generational family history and tumour ER and PR status. Predictors of mutation status were identified using multiple logistic regression. Areas under receiver operator characteristic (ROC) curves were estimated using five-fold stratified cross-validation. RESULTS: The probability of being a BRCA1 mutation carrier increased with number of selected histology features even after adjusting for family history and ER and PR status (P<0.0001). From the most parsimonious multivariate model, the odds ratio for being a carrier were: 9.7 (95% confidence interval: 2.6-47.0) for trabecular growth pattern (P=0.001); 7.8 (2.7-25.7) for mitotic index over 50 mitoses per 10 high-powered field (P=0.0003); and 2.7 (1.3-5.9) for each first-degree relative with breast cancer diagnosed before age 60 years (P=0.01).The area under the ROC curve was 0.87 (0.83-0.90). CONCLUSION: Pathology review, with attention to a few specific morphological features of invasive breast cancers, can identify almost all BRCA1 germline mutation carriers among women with early-onset breast cancer without taking into account family history.
  • Item
    Thumbnail Image
    Whole Exome Sequencing Suggests Much of Non-BRCA1/BRCA2 Familial Breast Cancer Is Due to Moderate and Low Penetrance Susceptibility Alleles
    Javier Gracia-Aznarez, F ; Fernandez, V ; Pita, G ; Peterlongo, P ; Dominguez, O ; de la Hoya, M ; Duran, M ; Osorio, A ; Moreno, L ; Gonzalez-Neira, A ; Manuel Rosa-Rosa, J ; Sinilnikova, O ; Mazoyer, S ; Hopper, J ; Lazaro, C ; Southey, M ; Odefrey, F ; Manoukian, S ; Catucci, I ; Caldes, T ; Lynch, HT ; Hilbers, FSM ; van Asperen, CJ ; Vasen, HFA ; Goldgar, D ; Radice, P ; Devilee, P ; Benitez, J ; Toland, AE (PUBLIC LIBRARY SCIENCE, 2013-02-08)
    The identification of the two most prevalent susceptibility genes in breast cancer, BRCA1 and BRCA2, was the beginning of a sustained effort to uncover new genes explaining the missing heritability in this disease. Today, additional high, moderate and low penetrance genes have been identified in breast cancer, such as P53, PTEN, STK11, PALB2 or ATM, globally accounting for around 35 percent of the familial cases. In the present study we used massively parallel sequencing to analyze 7 BRCA1/BRCA2 negative families, each having at least 6 affected women with breast cancer (between 6 and 10) diagnosed under the age of 60 across generations. After extensive filtering, Sanger sequencing validation and co-segregation studies, variants were prioritized through either control-population studies, including up to 750 healthy individuals, or case-control assays comprising approximately 5300 samples. As a result, a known moderate susceptibility indel variant (CHEK2 1100delC) and a catalogue of 11 rare variants presenting signs of association with breast cancer were identified. All the affected genes are involved in important cellular mechanisms like DNA repair, cell proliferation and survival or cell cycle regulation. This study highlights the need to investigate the role of rare variants in familial cancer development by means of novel high throughput analysis strategies optimized for genetically heterogeneous scenarios. Even considering the intrinsic limitations of exome resequencing studies, our findings support the hypothesis that the majority of non-BRCA1/BRCA2 breast cancer families might be explained by the action of moderate and/or low penetrance susceptibility alleles.
  • Item
    Thumbnail Image
    A PALB2 mutation associated with high risk of breast cancer
    Southey, MC ; Teo, ZL ; Dowty, JG ; Odefrey, FA ; Park, DJ ; Tischkowitz, M ; Sabbaghian, N ; Apicella, C ; Byrnes, GB ; Winship, I ; Baglietto, L ; Giles, GG ; Goldgar, DE ; Foulkes, WD ; Hopper, JL (BMC, 2010)
    NTRODUCTION: As a group, women who carry germline mutations in partner and localizer of breast cancer 2 susceptibility protein (PALB2) are at increased risk of breast cancer. Little is known about by how much or whether risk differs by mutation or family history, owing to the paucity of studies of cases unselected for family history. METHODS: We screened 1,403 case probands for PALB2 mutations in a population-based study of Australian women with invasive breast cancer stratified by age at onset. The age-specific risk of breast cancer was estimated from the cancer histories of first- and second-degree relatives of mutation-carrying probands using a modified segregation analysis that included a polygenic modifier and was conditioned on the carrier case proband. Further screening for PALB2 c.3113G > A (W1038X) was conducted for 779 families with multiple cases of breast cancer ascertained through family cancer clinics in Australia and New Zealand and 764 population-based controls. RESULTS: We found five independent case probands in the population-based sample with the protein-truncating mutation PALB2 c.3113G > A (W1038X); 2 of 695 were diagnosed before age 40 years and 3 of 708 were diagnosed when between ages 40 and 59 years. Both of the two early-onset carrier case probands had very strong family histories of breast cancer. Further testing found that the mutation segregated with breast cancer in these families. No c.3113G > A (W1038X) carriers were found in 764 population-based unaffected controls. The hazard ratio was estimated to be 30.1 (95% confidence interval (CI), 7.5 to 120; P < 0.0001), and the corresponding cumulative risk estimates were 49% (95% CI, 15 to 93) to age 50 and 91% (95% CI, 44 to 100) to age 70. We found another eight families carrying this mutation in 779 families with multiple cases of breast cancer ascertained through family cancer clinics. CONCLUSIONS: The PALB2 c.3113G > A mutation appears to be associated with substantial risks of breast cancer that are of clinical relevance.
  • Item
    Thumbnail Image
    Contribution of large genomic BRCA1 alterations to early-onset breast cancer selected for family history and tumour morphology: a report from The Breast Cancer Family Registry
    Smith, LD ; Tesoriero, AA ; Wong, EM ; Ramus, SJ ; O'Malley, FP ; Mulligan, AM ; Terry, MB ; Senie, RT ; Santella, RM ; John, EM ; Andrulis, IL ; Ozcelik, H ; Daly, MB ; Godwin, AK ; Buys, SS ; Fox, S ; Goldgar, DE ; Giles, GG ; Hopper, JL ; Southey, MC (BIOMED CENTRAL LTD, 2011)
    INTRODUCTION: Selecting women affected with breast cancer who are most likely to carry a germline mutation in BRCA1 and applying the most appropriate test methodology remains challenging for cancer genetics services. We sought to test the value of selecting women for BRCA1 mutation testing on the basis of family history and/or breast tumour morphology criteria as well as the value of testing for large genomic alterations in BRCA1. METHODS: We studied women participating in the Breast Cancer Family Registry (BCFR), recruited via population-based sampling, who had been diagnosed with breast cancer before the age of 40 years who had a strong family history of breast or ovarian cancer (n = 187) and/or a first primary breast tumour with morphological features consistent with carrying a BRCA1 germline mutation (n = 133; 37 met both criteria). An additional 184 women diagnosed before the age of 40 years who had a strong family history of breast or ovarian cancer and who were not known to carry a germline BRCA1 mutation were selected from among women who had been recruited into the BCFR from clinical genetics services. These 467 women had been screened for BRCA1 germline mutations, and we expanded this testing to include a screen for large genomic BRCA1 alterations using Multiplex Ligation-dependent Probe Amplification. RESULTS: Twelve large genomic BRCA1 alterations were identified, including 10 (4%) of the 283 women selected from among the population-based sample. In total, 18 (12%), 18 (19%) and 16 (43%) BRCA1 mutations were identified in the population-based groups selected on the basis of family history only (n = 150), the group selected on the basis of tumour morphology only (n = 96) and meeting both criteria (n = 37), respectively. CONCLUSIONS: Large genomic alterations accounted for 19% of all BRCA1 mutations identified. This study emphasises the value of combining information about family history, age at diagnosis and tumour morphology when selecting women for germline BRCA1 mutation testing as well as including a screen for large genomic alterations.
  • Item
    Thumbnail Image
    RAD51B in Familial Breast Cancer
    Pelttari, LM ; Khan, S ; Vuorela, M ; Kiiski, JI ; Vilske, S ; Nevanlinna, V ; Ranta, S ; Schleutker, J ; Winqvist, R ; Kallioniemi, A ; Doerk, T ; Bogdanova, NV ; Figueroa, J ; Pharoah, PDP ; Schmidt, MK ; Dunning, AM ; Garcia-Closas, M ; Bolla, MK ; Dennis, J ; Michailidou, K ; Wang, Q ; Hopper, JL ; Southey, MC ; Rosenberg, EH ; Fasching, PA ; Beckmann, MW ; Peto, J ; dos-Santos-Silva, I ; Sawyer, EJ ; Tomlinson, I ; Burwinkel, B ; Surowy, H ; Guenel, P ; Truong, T ; Bojesen, SE ; Nordestgaard, BG ; Benitez, J ; Gonzalez-Neira, A ; Neuhausen, SL ; Anton-Culver, H ; Brenner, H ; Arndt, V ; Meindl, A ; Schmutzler, RK ; Brauch, H ; Bruening, T ; Lindblom, A ; Margolin, S ; Mannermaa, A ; Hartikainen, JM ; Chenevix-Trench, G ; Van Dyck, L ; Janssen, H ; Chang-Claude, J ; Rudolph, A ; Radice, P ; Peterlongo, P ; Hallberg, E ; Olson, JE ; Giles, GG ; Milne, RL ; Haiman, CA ; Schumacher, F ; Simard, J ; Dumont, M ; Kristensen, V ; Borresen-Dale, A-L ; Zheng, W ; Beeghly-Fadiel, A ; Grip, M ; Andrulis, IL ; Glendon, G ; Devilee, P ; Seynaeve, C ; Hooning, MJ ; Collee, M ; Cox, A ; Cross, SS ; Shah, M ; Luben, RN ; Hamann, U ; Torres, D ; Jakubowska, A ; Lubinski, J ; Couch, FJ ; Yannoukakos, D ; Orr, N ; Swerdlow, A ; Darabi, H ; Li, J ; Czene, K ; Hall, P ; Easton, DF ; Mattson, J ; Blomqvist, C ; Aittomaki, K ; Nevanlinna, H ; Brusgaard, K (PUBLIC LIBRARY SCIENCE, 2016-05-05)
    Common variation on 14q24.1, close to RAD51B, has been associated with breast cancer: rs999737 and rs2588809 with the risk of female breast cancer and rs1314913 with the risk of male breast cancer. The aim of this study was to investigate the role of RAD51B variants in breast cancer predisposition, particularly in the context of familial breast cancer in Finland. We sequenced the coding region of RAD51B in 168 Finnish breast cancer patients from the Helsinki region for identification of possible recurrent founder mutations. In addition, we studied the known rs999737, rs2588809, and rs1314913 SNPs and RAD51B haplotypes in 44,791 breast cancer cases and 43,583 controls from 40 studies participating in the Breast Cancer Association Consortium (BCAC) that were genotyped on a custom chip (iCOGS). We identified one putatively pathogenic missense mutation c.541C>T among the Finnish cancer patients and subsequently genotyped the mutation in additional breast cancer cases (n = 5259) and population controls (n = 3586) from Finland and Belarus. No significant association with breast cancer risk was seen in the meta-analysis of the Finnish datasets or in the large BCAC dataset. The association with previously identified risk variants rs999737, rs2588809, and rs1314913 was replicated among all breast cancer cases and also among familial cases in the BCAC dataset. The most significant association was observed for the haplotype carrying the risk-alleles of all the three SNPs both among all cases (odds ratio (OR): 1.15, 95% confidence interval (CI): 1.11-1.19, P = 8.88 x 10-16) and among familial cases (OR: 1.24, 95% CI: 1.16-1.32, P = 6.19 x 10-11), compared to the haplotype with the respective protective alleles. Our results suggest that loss-of-function mutations in RAD51B are rare, but common variation at the RAD51B region is significantly associated with familial breast cancer risk.
  • Item
    Thumbnail Image
    Somatic mutations of the coding microsatellites within the beta-2-microglobulin gene in mismatch repair-deficient colorectal cancers and adenomas
    Clendenning, M ; Huang, A ; Jayasekara, H ; Lorans, M ; Preston, S ; O'Callaghan, N ; Pope, BJ ; Macrae, FA ; Winship, IM ; Milne, RL ; Giles, GG ; English, DR ; Hopper, JL ; Win, AK ; Jenkins, MA ; Southey, MC ; Rosty, C ; Buchanan, DD (SPRINGER, 2018-01)
    In colorectal cancers (CRCs) with tumour mismatch repair (MMR) deficiency, genes involved in the host immune response that contain microsatellites in their coding regions, including beta-2-microglobulin (B2M), can acquire mutations that may alter the immune response, tumour progression and prognosis. We screened the coding microsatellites within B2M for somatic mutations in MMR-deficient CRCs and adenomas to determine associations with tumour subtypes, clinicopathological features and survival. Incident MMR-deficient CRCs from Australasian Colorectal Cancer Family Registry (ACCFR) and the Melbourne Collaborative Cohort Study participants (n = 144) and 63 adenomas from 41 MMR gene mutation carriers from the ACCFR were screened for somatic mutations within five coding microsatellites of B2M. Hazard ratios (HR) and 95% confidence intervals (CI) for overall survival by B2M mutation status were estimated using Cox regression, adjusting for age at CRC diagnosis, sex, AJCC stage and grade. B2M mutations occurred in 30 (20.8%) of the 144 MMR-deficient CRCs (29% of the MLH1-methylated, 17% of the Lynch syndrome and 9% of the suspected Lynch CRCs). No B2M mutations were identified in the 63 adenomas tested. B2M mutations differed by site, stage, grade and lymphocytic infiltration although none reached statistical significance (p > 0.05). The HR for overall survival for B2M mutated CRC was 0.65 (95% CI 0.29-1.48) compared with B2M wild-type. We observed differences in B2M mutation status in MMR-deficient CRC by tumour subtypes, site, stage, grade, immune infiltrate and for overall survival that warrant further investigation in larger studies before B2M mutation status can be considered to have clinical utility.
  • Item
    No Preview Available
    Fine-Scale Mapping of the 4q24 Locus Identifies Two Independent Loci Associated with Breast Cancer Risk
    Guo, X ; Long, J ; Zeng, C ; Michailidou, K ; Ghoussaini, M ; Bolla, MK ; Wang, Q ; Milne, RL ; Shu, X-O ; Cai, Q ; Beesley, J ; Kar, SP ; Andrulis, IL ; Anton-Culver, H ; Arndt, V ; Beckmann, MW ; Beeghly-Fadiel, A ; Benitez, J ; Blot, W ; Bogdanova, N ; Bojesen, SE ; Brauch, H ; Brenner, H ; Brinton, L ; Broeks, A ; Bruening, T ; Burwinkel, B ; Cai, H ; Canisius, S ; Chang-Claude, J ; Choi, J-Y ; Couch, FJ ; Cox, A ; Cross, SS ; Czene, K ; Darabi, H ; Devilee, P ; Droit, A ; Doerk, T ; Fasching, PA ; Fletcher, O ; Flyger, H ; Fostira, F ; Gaborieau, V ; Garcia-Closas, M ; Giles, GG ; Grip, M ; Guenel, P ; Haiman, CA ; Hamann, U ; Hartman, M ; Hollestelle, A ; Hopper, JL ; Hsiung, C-N ; Ito, H ; Jakubowska, A ; Johnson, N ; Kabisch, M ; Kang, D ; Khan, S ; Knight, JA ; Kosma, V-M ; Lambrechts, D ; Le Marchand, L ; Li, J ; Lindblom, A ; Lophatananon, A ; Lubinski, J ; Mannermaa, A ; Manoukian, S ; Margolin, S ; Marme, F ; Matsuo, K ; McLean, CA ; Meindl, A ; Muir, K ; Neuhausen, SL ; Nevanlinna, H ; Nord, S ; Olson, JE ; Orr, N ; Peterlongo, P ; Putti, TC ; Rudolph, A ; Sangrajrang, S ; Sawyer, EJ ; Schmidt, MK ; Schmutzler, RK ; Shen, C-Y ; Shi, J ; Shrubsole, MJ ; Southey, MC ; Swerdlow, A ; Teo, SH ; Thienpont, B ; Toland, AE ; Tollenaar, RAEM ; Tomlinson, IPM ; Truong, T ; Tseng, C-C ; van den Ouweland, A ; Wen, W ; Winqvist, R ; Wu, A ; Yip, CH ; Zamora, MP ; Zheng, Y ; Hall, P ; Pharoah, PDP ; Simard, J ; Chenevix-Trench, G ; Dunning, AM ; Easton, DF ; Zheng, W (AMER ASSOC CANCER RESEARCH, 2015-11)
    BACKGROUND: A recent association study identified a common variant (rs9790517) at 4q24 to be associated with breast cancer risk. Independent association signals and potential functional variants in this locus have not been explored. METHODS: We conducted a fine-mapping analysis in 55,540 breast cancer cases and 51,168 controls from the Breast Cancer Association Consortium. RESULTS: Conditional analyses identified two independent association signals among women of European ancestry, represented by rs9790517 [conditional P = 2.51 × 10(-4); OR, 1.04; 95% confidence interval (CI), 1.02-1.07] and rs77928427 (P = 1.86 × 10(-4); OR, 1.04; 95% CI, 1.02-1.07). Functional annotation using data from the Encyclopedia of DNA Elements (ENCODE) project revealed two putative functional variants, rs62331150 and rs73838678 in linkage disequilibrium (LD) with rs9790517 (r(2) ≥ 0.90) residing in the active promoter or enhancer, respectively, of the nearest gene, TET2. Both variants are located in DNase I hypersensitivity and transcription factor-binding sites. Using data from both The Cancer Genome Atlas (TCGA) and Molecular Taxonomy of Breast Cancer International Consortium (METABRIC), we showed that rs62331150 was associated with level of expression of TET2 in breast normal and tumor tissue. CONCLUSION: Our study identified two independent association signals at 4q24 in relation to breast cancer risk and suggested that observed association in this locus may be mediated through the regulation of TET2. IMPACT: Fine-mapping study with large sample size warranted for identification of independent loci for breast cancer risk.
  • Item
    No Preview Available
    Alcohol consumption for different periods in life, intake pattern over time and all-cause mortality
    Jayasekara, H ; MacInnis, RJ ; Hodge, AM ; Hopper, JL ; Giles, GG ; Room, R ; English, DR (OXFORD UNIV PRESS, 2015-12)
    BACKGROUND: Conventionally, cohort studies have assessed the association between alcohol and all-cause mortality by using alcohol intake at enrolment. METHODS: In the Melbourne Collaborative Cohort Study, participants were asked about usual frequency and quantity of beverage-specific alcohol intake for 10-year periods starting at age 20 from which current, past and lifetime intakes were calculated. We used Cox regression to estimate hazard ratios for mortality for 39 577 participants of the Melbourne Collaborative Cohort Study aged 40-69 at baseline. RESULTS: After a mean follow-up of 15 years/person, we identified 4639 deaths. Associations between all-cause mortality and lifetime, current (baseline) and past intake were J shaped, with lower mortality at low intake (e.g. <40 g/day for men and 10 g/day for women using lifetime intake) and elevated mortality at higher intake. For men, consistent light-to-moderate drinking (>0-39/>0-39 g/day) from age 20 to baseline age was associated with a 16% lower mortality, while heavy drinking at both ages (≥80/≥40 and ≥40/0 g/day) was associated with higher mortality compared with stable abstinence. CONCLUSIONS: Our findings support a reduced mortality risk associated with low-dose drinking but also highlight a higher mortality risk for consistent heavy drinking from a young age.
  • Item
    No Preview Available
    No evidence that protein truncating variants in BRIP1 are associated with breast cancer risk: implications for gene panel testing
    Easton, DF ; Lesueur, F ; Decker, B ; Michailidou, K ; Li, J ; Allen, J ; Luccarini, C ; Pooley, KA ; Shah, M ; Bolla, MK ; Wang, Q ; Dennis, J ; Ahmad, J ; Thompson, ER ; Damiola, F ; Pertesi, M ; Voegele, C ; Mebirouk, N ; Robinot, N ; Durand, G ; Forey, N ; Luben, RN ; Ahmed, S ; Aittomaki, K ; Anton-Culver, H ; Arndt, V ; Baynes, C ; Beckman, MW ; Benitez, J ; Van Den Berg, D ; Blot, WJ ; Bogdanova, NV ; Bojesen, SE ; Brenner, H ; Chang-Claude, J ; Chia, KS ; Choi, J-Y ; Conroy, DM ; Cox, A ; Cross, SS ; Czene, K ; Darabi, H ; Devilee, P ; Eriksson, M ; Fasching, PA ; Figueroa, J ; Flyger, H ; Fostira, F ; Garcia-Closas, M ; Giles, GG ; Glendon, G ; Gonzalez-Neira, A ; Guenel, P ; Haiman, CA ; Hall, P ; Hart, SN ; Hartman, M ; Hooning, MJ ; Hsiung, C-N ; Ito, H ; Jakubowska, A ; James, PA ; John, EM ; Johnson, N ; Jones, M ; Kabisch, M ; Kang, D ; Kosma, V-M ; Kristensen, V ; Lambrechts, D ; Li, N ; Lindblom, A ; Long, J ; Lophatananon, A ; Lubinski, J ; Mannermaa, A ; Manoukian, S ; Margolin, S ; Matsuo, K ; Meindl, A ; Mitchell, G ; Muir, K ; Nevelsteen, I ; van den Ouweland, A ; Peterlongo, P ; Phuah, SY ; Pylkas, K ; Rowley, SM ; Sangrajrang, S ; Schmutzler, RK ; Shen, C-Y ; Shu, X-O ; Southey, MC ; Surowy, H ; Swerdlow, A ; Teo, SH ; Tollenaar, RAEM ; Tomlinson, I ; Torres, D ; Truong, T ; Vachon, C ; Verhoef, S ; Wong-Brown, M ; Zheng, W ; Zheng, Y ; Nevanlinna, H ; Scott, RJ ; Andrulis, IL ; Wu, AH ; Hopper, JL ; Couch, FJ ; Winqvist, R ; Burwinkel, B ; Sawyer, EJ ; Schmidt, MK ; Rudolph, A ; Doerk, T ; Brauch, H ; Hamann, U ; Neuhausen, SL ; Milne, RL ; Fletcher, O ; Pharoah, PDP ; Campbell, IG ; Dunning, AM ; Le Calvez-Kelm, F ; Goldgar, DE ; Tavtigian, SV ; Chenevix-Trench, G (BMJ PUBLISHING GROUP, 2016-05)
    BACKGROUND: BRCA1 interacting protein C-terminal helicase 1 (BRIP1) is one of the Fanconi Anaemia Complementation (FANC) group family of DNA repair proteins. Biallelic mutations in BRIP1 are responsible for FANC group J, and previous studies have also suggested that rare protein truncating variants in BRIP1 are associated with an increased risk of breast cancer. These studies have led to inclusion of BRIP1 on targeted sequencing panels for breast cancer risk prediction. METHODS: We evaluated a truncating variant, p.Arg798Ter (rs137852986), and 10 missense variants of BRIP1, in 48 144 cases and 43 607 controls of European origin, drawn from 41 studies participating in the Breast Cancer Association Consortium (BCAC). Additionally, we sequenced the coding regions of BRIP1 in 13 213 cases and 5242 controls from the UK, 1313 cases and 1123 controls from three population-based studies as part of the Breast Cancer Family Registry, and 1853 familial cases and 2001 controls from Australia. RESULTS: The rare truncating allele of rs137852986 was observed in 23 cases and 18 controls in Europeans in BCAC (OR 1.09, 95% CI 0.58 to 2.03, p=0.79). Truncating variants were found in the sequencing studies in 34 cases (0.21%) and 19 controls (0.23%) (combined OR 0.90, 95% CI 0.48 to 1.70, p=0.75). CONCLUSIONS: These results suggest that truncating variants in BRIP1, and in particular p.Arg798Ter, are not associated with a substantial increase in breast cancer risk. Such observations have important implications for the reporting of results from breast cancer screening panels.
  • Item
    Thumbnail Image
    Association of breast cancer risk with genetic variants showing differential allelic expression: Identification of a novel breast cancer susceptibility locus at 4q21
    Hamdi, Y ; Soucy, P ; Adoue, V ; Michailidou, K ; Canisius, S ; Lemacon, A ; Droit, A ; Andrulis, IL ; Anton-Culver, H ; Arndt, V ; Baynes, C ; Blomqvist, C ; Bogdanova, NV ; Bojesen, SE ; Bolla, MK ; Bonanni, B ; Borresen-Dale, A-L ; Brand, JS ; Brauch, H ; Brenner, H ; Broeks, A ; Burwinkel, B ; Chang-Claude, J ; Couch, FJ ; Cox, A ; Cross, SS ; Czene, K ; Darabi, H ; Dennis, J ; Devilee, P ; Doerk, T ; Dos-Santos-Silva, I ; Eriksson, M ; Fasching, PA ; Figueroa, J ; Flyger, H ; Garcia-Closas, M ; Giles, GG ; Goldberg, MS ; Gonzalez-Neira, A ; Grenaker-Alnaes, G ; Guenel, P ; Haeberle, L ; Haiman, CA ; Hamann, U ; Hallberg, E ; Hooning, MJ ; Hopper, JL ; Jakubowska, A ; Jones, M ; Kabisch, M ; Kataja, V ; Lambrechts, D ; Le Marchand, L ; Lindblom, A ; Lubinski, J ; Mannermaa, A ; Maranian, M ; Margolin, S ; Marme, F ; Milne, RL ; Neuhausen, SL ; Nevanlinna, H ; Neven, P ; Olswold, C ; Peto, J ; Plaseska-Karanfilska, D ; Pylkaes, K ; Radice, P ; Rudolph, A ; Sawyer, EJ ; Schmidt, MK ; Shu, X-O ; Southey, MC ; Swerdlow, A ; Tollenaar, RAEM ; Tomlinson, I ; Torres, D ; Truong, T ; Vachon, C ; Van Den Ouweland, AMW ; Wang, Q ; Winqvist, R ; Zheng, W ; Benitez, J ; Chenevix-Trench, G ; Dunning, AM ; Pharoah, PDP ; Kristensen, V ; Hall, P ; Easton, DF ; Pastinen, T ; Nord, S ; Simard, J (IMPACT JOURNALS LLC, 2016-12-06)
    There are significant inter-individual differences in the levels of gene expression. Through modulation of gene expression, cis-acting variants represent an important source of phenotypic variation. Consequently, cis-regulatory SNPs associated with differential allelic expression are functional candidates for further investigation as disease-causing variants. To investigate whether common variants associated with differential allelic expression were involved in breast cancer susceptibility, a list of genes was established on the basis of their involvement in cancer related pathways and/or mechanisms. Thereafter, using data from a genome-wide map of allelic expression associated SNPs, 313 genetic variants were selected and their association with breast cancer risk was then evaluated in 46,451 breast cancer cases and 42,599 controls of European ancestry ascertained from 41 studies participating in the Breast Cancer Association Consortium. The associations were evaluated with overall breast cancer risk and with estrogen receptor negative and positive disease. One novel breast cancer susceptibility locus on 4q21 (rs11099601) was identified (OR = 1.05, P = 5.6x10-6). rs11099601 lies in a 135 kb linkage disequilibrium block containing several genes, including, HELQ, encoding the protein HEL308 a DNA dependant ATPase and DNA Helicase involved in DNA repair, MRPS18C encoding the Mitochondrial Ribosomal Protein S18C and FAM175A (ABRAXAS), encoding a BRCA1 BRCT domain-interacting protein involved in DNA damage response and double-strand break (DSB) repair. Expression QTL analysis in breast cancer tissue showed rs11099601 to be associated with HELQ (P = 8.28x10-14), MRPS18C (P = 1.94x10-27) and FAM175A (P = 3.83x10-3), explaining about 20%, 14% and 1%, respectively of the variance inexpression of these genes in breast carcinomas.