118
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Heterogeneity of Breast Cancer Associations with Five Susceptibility Loci by Clinical and Pathological Characteristics

      research-article
      1 , * , 2 , 3 , 4 , 4 , 1 , 5 , 6 , 5 , 7 , 8 , 9 , 8 , 8 , 8 , 8 , 10 , 11 , 11 , 12 , 13 , 14 , 15 , 16 , 16 , 16 , 16 , 17 , 17 , 17 , 3 , 3 , 18 , 19 , 20 , 18 , 21 , 22 , 23 , 24 , 25 , 26 , 21 , 22 , 25 , 26 , 27 , 27 , 27 , 27 , 27 , 28 , 28 , 29 , 30 , 31 , 32 , 33 , 33 , 34 , 34 , 34 , 35 , 35 , 36 , 37 , 38 , 38 , 38 , 38 , 38 , 39 , 39 , 39 , 40 , 40 , 41 , 42 , 42 , 43 , 43 , 44 , 45 , 46 , 46 , 46 , 47 , 46 , 47 , 1 , 48 , 49 , 1 , 50 , 51 , 51 , 52 , 52 , 52 , 53 , 54 , 54 , 54 , 2 , 2 , 55 , 56 , 57 , 56 , 4 , 4 , 4 , 4 , 58 , 59 , 60 , 60 , 60 , 60 , 1 , 1 , 61 , 62 , 4 , 4
      PLoS Genetics
      Public Library of Science

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          A three-stage genome-wide association study recently identified single nucleotide polymorphisms (SNPs) in five loci (fibroblast growth receptor 2 ( FGFR2), trinucleotide repeat containing 9 ( TNRC9), mitogen-activated protein kinase 3 K1 ( MAP3K1), 8q24, and lymphocyte-specific protein 1 ( LSP1)) associated with breast cancer risk. We investigated whether the associations between these SNPs and breast cancer risk varied by clinically important tumor characteristics in up to 23,039 invasive breast cancer cases and 26,273 controls from 20 studies. We also evaluated their influence on overall survival in 13,527 cases from 13 studies. All participants were of European or Asian origin. rs2981582 in FGFR2 was more strongly related to ER-positive (per-allele OR (95%CI) = 1.31 (1.27–1.36)) than ER-negative (1.08 (1.03–1.14)) disease (P for heterogeneity = 10 −13). This SNP was also more strongly related to PR-positive, low grade and node positive tumors (P = 10 −5, 10 −8, 0.013, respectively). The association for rs13281615 in 8q24 was stronger for ER-positive, PR-positive, and low grade tumors (P = 0.001, 0.011 and 10 −4, respectively). The differences in the associations between SNPs in FGFR2 and 8q24 and risk by ER and grade remained significant after permutation adjustment for multiple comparisons and after adjustment for other tumor characteristics. Three SNPs (rs2981582, rs3803662, and rs889312) showed weak but significant associations with ER-negative disease, the strongest association being for rs3803662 in TNRC9 (1.14 (1.09–1.21)). rs13281615 in 8q24 was associated with an improvement in survival after diagnosis (per-allele HR = 0.90 (0.83–0.97). The association was attenuated and non-significant after adjusting for known prognostic factors. Our findings show that common genetic variants influence the pathological subtype of breast cancer and provide further support for the hypothesis that ER-positive and ER-negative disease are biologically distinct. Understanding the etiologic heterogeneity of breast cancer may ultimately result in improvements in prevention, early detection, and treatment.

          Author Summary

          This report from the Breast Cancer Association Consortium evaluates whether common variants in five recently identified breast cancer susceptibility loci ( FGFR2, TNRC9, MAP3K1, 8q24, and LSP1) influence the clinical presentation of breast cancer and survival after diagnosis. We studied these susceptibility loci in relation to clinically important tumor characteristics in up to 23,039 invasive breast cancer cases and 26,273 controls of European or Asian origin from 20 studies. The association, with overall survival, was evaluated in 13,527 cases from 13 studies. The most notable findings were that the genetic variants in the fibroblast growth factor receptor 2 ( FGFR2) gene and the 8q24 region were more strongly related to ER-positive than ER-negative disease, and to low rather than high grade tumors. The loci did not significantly influence survival after accounting for known prognostic factors. Analyses indicated that common genetic variants influence the pathological subtype of breast cancer and provide further support for the hypothesis that ER-positive and ER-negative diseases are biologically distinct tumors. Understanding the etiologic heterogeneity of breast cancer may ultimately result in improvements in prevention, early detection, and treatment.

          Related collections

          Most cited references27

          • Record: found
          • Abstract: found
          • Article: not found

          A genome-wide association study identifies alleles in FGFR2 associated with risk of sporadic postmenopausal breast cancer.

          We conducted a genome-wide association study (GWAS) of breast cancer by genotyping 528,173 SNPs in 1,145 postmenopausal women of European ancestry with invasive breast cancer and 1,142 controls. We identified four SNPs in intron 2 of FGFR2 (which encodes a receptor tyrosine kinase and is amplified or overexpressed in some breast cancers) that were highly associated with breast cancer and confirmed this association in 1,776 affected individuals and 2,072 controls from three additional studies. Across the four studies, the association with all four SNPs was highly statistically significant (P(trend) for the most strongly associated SNP (rs1219648) = 1.1 x 10(-10); population attributable risk = 16%). Four SNPs at other loci most strongly associated with breast cancer in the initial GWAS were not associated in the replication studies. Our summary results from the GWAS are available online in a form that should speed the identification of additional risk loci.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Genome-wide association study of prostate cancer identifies a second risk locus at 8q24.

            Recently, common variants on human chromosome 8q24 were found to be associated with prostate cancer risk. While conducting a genome-wide association study in the Cancer Genetic Markers of Susceptibility project with 550,000 SNPs in a nested case-control study (1,172 cases and 1,157 controls of European origin), we identified a new association at 8q24 with an independent effect on prostate cancer susceptibility. The most significant signal is 70 kb centromeric to the previously reported SNP, rs1447295, but shows little evidence of linkage disequilibrium with it. A combined analysis with four additional studies (total: 4,296 cases and 4,299 controls) confirms association with prostate cancer for rs6983267 in the centromeric locus (P = 9.42 x 10(-13); heterozygote odds ratio (OR): 1.26, 95% confidence interval (c.i.): 1.13-1.41; homozygote OR: 1.58, 95% c.i.: 1.40-1.78). Each SNP remained significant in a joint analysis after adjusting for the other (rs1447295 P = 1.41 x 10(-11); rs6983267 P = 6.62 x 10(-10)). These observations, combined with compelling evidence for a recombination hotspot between the two markers, indicate the presence of at least two independent loci within 8q24 that contribute to prostate cancer in men of European ancestry. We estimate that the population attributable risk of the new locus, marked by rs6983267, is higher than the locus marked by rs1447295 (21% versus 9%).
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              A genome-wide association scan of tag SNPs identifies a susceptibility variant for colorectal cancer at 8q24.21.

              Much of the variation in inherited risk of colorectal cancer (CRC) is probably due to combinations of common low risk variants. We conducted a genome-wide association study of 550,000 tag SNPs in 930 familial colorectal tumor cases and 960 controls. The most strongly associated SNP (P = 1.72 x 10(-7), allelic test) was rs6983267 at 8q24.21. To validate this finding, we genotyped rs6983267 in three additional CRC case-control series (4,361 affected individuals and 3,752 controls; 1,901 affected individuals and 1,079 controls; 1,072 affected individuals and 415 controls) and replicated the association, providing P = 1.27 x 10(-14) (allelic test) overall, with odds ratios (ORs) of 1.27 (95% confidence interval (c.i.): 1.16-1.39) and 1.47 (95% c.i.: 1.34-1.62) for heterozygotes and rare homozygotes, respectively. Analyses based on 1,477 individuals with colorectal adenoma and 2,136 controls suggest that susceptibility to CRC is mediated through development of adenomas (OR = 1.21, 95% c.i.: 1.10-1.34; P = 6.89 x 10(-5)). These data show that common, low-penetrance susceptibility alleles predispose to colorectal neoplasia.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                PLoS Genet
                plos
                plosgen
                PLoS Genetics
                Public Library of Science (San Francisco, USA )
                1553-7390
                1553-7404
                April 2008
                April 2008
                25 April 2008
                : 4
                : 4
                : e1000054
                Affiliations
                [1 ]Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Marylan, United States of America
                [2 ]Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
                [3 ]Department of Obstetrics and Gynaecology, Helsinki University Central Hospital, Helsinki, Finland
                [4 ]Department of Oncology, University of Cambridge, Cambridge, United Kingdom
                [5 ]Department of Clinical Biochemistry, Herlev and Bispebjerg University Hospitals, University of Copenhagen, Denmark
                [6 ]Department of Bispebjerg University Hospitals, University of Copenhagen, Denmark
                [7 ]Department of Breast Surgery, Herlev University Hospital, University of Copenhagen, Denmark
                [8 ]Spanish National Cancer Centre, Madrid, Spain
                [9 ]Monte Naranco Hospital, Oviedo, Spain
                [10 ]La Paz Hospital, Madrid, Spain
                [11 ]Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart and University of Tübingen, Tübingen, Germany
                [12 ]Deutsches Krebsforschungszentrum Heidelberg, Heidelberg, Germany
                [13 ]Evangelische Kliniken Bonn gGmhH Johanniter Krankenhaus, Bonn, Germany
                [14 ]Berufsgenossenschaftliches Forschungsinstitut für Arbeitsmedizin, Ruhr University Bochum, Germany
                [15 ]Institute für Pathology, University Bonn, Bonn, Germany
                [16 ]Department of Gynecology and Obstetrics, Hannover Medical School, Hannover, Germany
                [17 ]Department of Radiation Oncology, Hannover Medical School, Hannover, Germany
                [18 ]Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland
                [19 ]Department of Clinical Genetics, Helsinki University Central Hospital, Helsinki, Finland
                [20 ]Department of Surgery, Helsinki University Central Hospital, Helsinki, Finland
                [21 ]Institute of Clinical Medicine, Pathology and Forensic Medicine, Biocenter Kuopio, University of Kuopio, Kuopio, Finland
                [22 ]Department of Pathology, Kuopio University Hospital, Kuopio, Finland
                [23 ]Department of Public Health and Clinical Nutrition, Biocenter Kuopio, University of Kuopio, Kuopio, Finland
                [24 ]Department of Surgery, Kuopio University Hospital, Kuopio, Finland
                [25 ]Department of Oncology, Kuopio University Hospital, Kuopio, Finland
                [26 ]Department of Oncology, Vaasa Central Hospital, Vaasa, Finland
                [27 ]The Queensland Institute of Medical Research Post Office, Royal Brisbane Hospital, Herston, Queensland, Australia
                [28 ]Peter MacCallum Cancer Institute, East Melbourne, Victoria, Australia
                [29 ]Departments of Human Genetics and Pathology, Leiden University Medical Center, Leiden, The Netherlands
                [30 ]Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
                [31 ]Department of Medical Decision Making, Leiden University Medical Center, Leiden, The Netherlands
                [32 ]Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
                [33 ]Department of Medical Oncology, Family Cancer Clinic, Erasmus MC-Daniel den Hoed Cancer Center, Rotterdam, The Netherlands
                [34 ]Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
                [35 ]Institute for Medical Biometrics and Epidemiology, University Clinic Hamburg-Eppendorf, Hamburg, Germany
                [36 ]Bioglobe GmbH, Hamburg, Germany
                [37 ]Molecular Biology Laboratory, Department of Obstetrics and Gynecology, University of Ulm, Ulm, Germany
                [38 ]Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
                [39 ]Cancer Epidemiology Centre, The Cancer Council Victoria, Melbourne, Victoria, Australia
                [40 ]Centre for MEGA Epidemiology, The University of Melbourne, Melbourne, Victoria, Australia
                [41 ]Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Melbourne, Victoria, Australia
                [42 ]Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
                [43 ]Epidemiology Program, Cancer Research Center of Hawaii, University of Hawaii, Honolulu, Hawaii, United States of America
                [44 ]Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
                [45 ]Program in Molecular and Genetic Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
                [46 ]Channing Laboratory, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
                [47 ]Program in Molecular and Genetic Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
                [48 ]Advanced Technology Center, National Cancer Institute, Gaithersburg, Maryland, United States of America
                [49 ]Department of Cancer Epidemiology and Prevention, Cancer Center and M. Sklodowska-Curie Institute of Oncology, Warsaw, Poland
                [50 ]Nofer Institute of Occupational Medicine, Lodz, Poland
                [51 ]Daniel den Hoed Cancer Center, Erasmus Medical Center, Department of Medical Oncology, Rotterdam, The Netherlands
                [52 ]Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
                [53 ]Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
                [54 ]Human Genetics, Genome Institute of Singapore, Singapore
                [55 ]Institute for Cancer Studies, Sheffield University Medical School, Sheffield, United Kingdom
                [56 ]Academic Unit of Surgical Oncology, Sheffield University Medical School, Sheffield, United Kingdom
                [57 ]Academic Unit of Pathology, Sheffield University Medical School, Sheffield, United Kingdom
                [58 ]Cancer Research UK, Cambridge Research Institute, Cambridge, United Kingdom
                [59 ]Molecular Epidemiology Unit, National Cancer Institute, Ratchathewi, Bangkok, Thailand
                [60 ]International Agency for Research on Cancer, Lyon, France
                [61 ]Office of Population Genomics, National Human Genome Research Institute, Bethesda, Maryland, United Stated of America
                [62 ]Environmental Health Sciences, University of Minnesota, Minneapolis, Minnesota, United States of America
                Baylor College of Medicine, United States of America
                Author notes

                Conceived and designed the experiments: M. Garcia-Closas, P. Hall, S. Bojesen, B. Nordestgaard, C. Axelsson, T. Dörk, J. Chang-Claude, D. Flesch-Janys, D. Hunter, S. Hankinson, D. Cox, R. Tamimi, P. Kraft, P. Brennan, J. McKay, D. Easton, P. Pharoah. Performed the experiments: K. Pooley, R. Milne, G. Ribas, A. González-Neira, J. Benítez, H. Brauch, C. Justenhoven, T. Dörk, P. Schürmann, N. Bogdanova, A. Mannermaa, J. Beesley, X. Chen, J. Chang-Claude, R. Salazar, S. Wang-Gohrke, F. Couch, M. Southey, D. Hunter, D. Cox, S. Chanock, J. Liu, A. Dunning, F. Odefrey, J. Struewing. Analyzed the data: M. Garcia-Closas, D. Richesson, T. Dörk, M. Tengström, V. Kataja, P. Huijts, S. Kropp, D. Flesch-Janys, P. Kraft, J. Tyrer, V. Gabrieau. Contributed reagents/materials/analysis tools: M. Garcia-Closas, P. Hall, H. Nevanlinna, K. Pooley, J. Morrison, S. Bojesen, B. Nordestgaard, C. Axelsson, H. Brauch, P. Hillemanns, M. Bremer, J. Karstens, R. Fagerholm, K. Aaltonen, K. Aittomäki, K. von Smitten, C. Blomqvist, M. Uusitupa, M. Eskelinen, V. Kosma, G. Chenevix-Trench, A. Spurdle, Australian Ovarian Cancer Management Group, The Kathleen Cuningham Foundation Consortium for Research into Familial Breast Cancer, P. Devilee, C. van Asperen, R. Tollenaar, J. Chang-Claude, D. Flesch-Janys, F. Couch, J. Olson, G. Giles, C. Haiman, B. Henderson, L. Kolonel, L. Le Marchand, D. Stram, A. Uitterlinden, J. Liu, K. Humphreys, K. Czene, J. Struewing. Wrote the paper: M. Garcia-Closas, P. Hall, H. Nevanlinna, P. Pharoah. Study design/data collection: M. Garcia-Closas, P. Hall, H. Nevanlinna, J. Morrison, J. Arias, P. Zamora, H. Brauch, C. Justenhoven, U. Hamann, Y. Ko, T. Bruening, S. Haas, R. Fagerholm, K. Aaltonen, A. Mannermaa, M. Uusitupa, M. Eskelinen, M. Tengström, V. Kosma, V. Kataja, G. Chenevix-Trench, A. Spurdle, C. van Asperen, C. Jacobi, R. Tollenaar, J. Klijn, T. Slanger, E. Mutschelknauss, F. Couch, E. Goode, J. Olson, C. Vachon, Z. Fredericksen, G. Giles, L. Baglietto, G. Severi, J. Hopper, D. English, M. Southey, C. Haiman, B. Henderson, L. Kolonel, L. Le Marchand, D. Stram, D. Hunter, S. Hankinson, R. Tamimi, M. Sherman, J. Lissowska, L. Brinton, B. Peplonska, J. Klijn, M. Hooning, H. Meijers-Heijboer, J. Collee, A. Ouweland, J. Liu, L. Lin, L. Yuqing, A. Cox, S. Balasubramanian, M. Reed, F. Blows, K. Driver, A. Dunning, B. Ponder, S. Sangrajrang, A. Sigurdson, M. Doody, B. Alexander, D. Easton, P. Pharoah. Pathology review: J. Arias, P. Zamora, S. Haas, M. Bremer, V. Kosma, M. Sherman, S. Cross, F. Blows, K. Driver.

                Article
                07-PLGE-RA-0661R5
                10.1371/journal.pgen.1000054
                2291027
                18437204
                7b8cdaff-1e29-4c12-9f15-8ed688d2edc2
                This is an open-access article distributed under the terms of the Creative Commons Public Domain declaration which stipulates that, once placed in the public domain, this work may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose.
                History
                : 16 August 2007
                : 18 March 2008
                Page count
                Pages: 10
                Categories
                Research Article
                Genetics and Genomics/Genetics of Disease
                Oncology/Breast Cancer

                Genetics
                Genetics

                Comments

                Comment on this article