Sir Peter MacCallum Department of Oncology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 7 of 7
  • Item
    No Preview Available
    COMPLEXO: identifying the missing heritability of breast cancer via next generation collaboration
    Southey, MC ; Park, DJ ; Nguyen-Dumont, T ; Campbell, I ; Thompson, E ; Trainer, AH ; Chenevix-Trench, G ; Simard, J ; Dumont, M ; Soucy, P ; Thomassen, M ; Jonson, L ; Pedersen, IS ; Hansen, TVO ; Nevanlinna, H ; Khan, S ; Sinilnikova, O ; Mazoyer, S ; Lesueur, F ; Damiola, F ; Schmutzler, R ; Meindl, A ; Hahnen, E ; Dufault, MR ; Chan, TC ; Kwong, A ; Barkardottir, R ; Radice, P ; Peterlongo, P ; Devilee, P ; Hilbers, F ; Benitez, J ; Kvist, A ; Torngren, T ; Easton, D ; Hunter, D ; Lindstrom, S ; Kraft, P ; Zheng, W ; Gao, Y-T ; Long, J ; Ramus, S ; Feng, B-J ; Weitzel, RN ; Nathanson, K ; Offit, K ; Joseph, V ; Robson, M ; Schrader, K ; Wang, SM ; Kim, YC ; Lynch, H ; Snyder, C ; Tavtigian, S ; Neuhausen, S ; Couch, FJ ; Goldgar, DE (BMC, 2013)
    Linkage analysis, positional cloning, candidate gene mutation scanning and genome-wide association study approaches have all contributed significantly to our understanding of the underlying genetic architecture of breast cancer. Taken together, these approaches have identified genetic variation that explains approximately 30% of the overall familial risk of breast cancer, implying that more, and likely rarer, genetic susceptibility alleles remain to be discovered.
  • Item
    No Preview Available
    GWAS meta-analysis and replication identifies three new susceptibility loci for ovarian cancer
    Pharoah, PDP ; Tsai, Y-Y ; Ramus, SJ ; Phelan, CM ; Goode, EL ; Lawrenson, K ; Buckley, M ; Fridley, BL ; Tyrer, JP ; Shen, H ; Weber, R ; Karevan, R ; Larson, MC ; Song, H ; Tessier, DC ; Bacot, F ; Vincent, D ; Cunningham, JM ; Dennis, J ; Dicks, E ; Aben, KK ; Anton-Culver, H ; Antonenkova, N ; Armasu, SM ; Baglietto, L ; Bandera, EV ; Beckmann, MW ; Birrer, MJ ; Bloom, G ; Bogdanova, N ; Brenton, JD ; Brinton, LA ; Brooks-Wilson, A ; Brown, R ; Butzow, R ; Campbell, I ; Carney, ME ; Carvalho, RS ; Chang-Claude, J ; Chen, YA ; Chen, Z ; Chow, W-H ; Cicek, MS ; Coetzee, G ; Cook, LS ; Cramer, DW ; Cybulski, C ; Dansonka-Mieszkowska, A ; Despierre, E ; Doherty, JA ; Doerk, T ; du Bois, A ; Duerst, M ; Eccles, D ; Edwards, R ; Ekici, AB ; Fasching, PA ; Fenstermacher, D ; Flanagan, J ; Gao, Y-T ; Garcia-Closas, M ; Gentry-Maharaj, A ; Giles, G ; Gjyshi, A ; Gore, M ; Gronwald, J ; Guo, Q ; Halle, MK ; Harter, P ; Hein, A ; Heitz, F ; Hillemanns, P ; Hoatlin, M ; Hogdall, E ; Hogdall, CK ; Hosono, S ; Jakubowska, A ; Jensen, A ; Kalli, KR ; Karlan, BY ; Kelemen, LE ; Kiemeney, LA ; Kjaer, SK ; Konecny, GE ; Krakstad, C ; Kupryjanczyk, J ; Lambrechts, D ; Lambrechts, S ; Le, ND ; Lee, N ; Lee, J ; Leminen, A ; Lim, BK ; Lissowska, J ; Lubinski, J ; Lundvall, L ; Lurie, G ; Massuger, LFAG ; Matsuo, K ; McGuire, V ; McLaughlin, JR ; Menon, U ; Modugno, F ; Moysich, KB ; Nakanishi, T ; Narod, SA ; Ness, RB ; Nevanlinna, H ; Nickels, S ; Noushmehr, H ; Odunsi, K ; Olson, S ; Orlow, I ; Paul, J ; Pejovic, T ; Pelttari, LM ; Permuth-Wey, J ; Pike, MC ; Poole, EM ; Qu, X ; Risch, HA ; Rodriguez-Rodriguez, L ; Rossing, MA ; Rudolph, A ; Runnebaum, I ; Rzepecka, IK ; Salvesen, HB ; Schwaab, I ; Severi, G ; Shen, H ; Shridhar, V ; Shu, X-O ; Sieh, W ; Southey, MC ; Spellman, P ; Tajima, K ; Teo, S-H ; Terry, KL ; Thompson, PJ ; Timorek, A ; Tworoger, SS ; van Altena, AM ; van den Berg, D ; Vergote, I ; Vierkant, RA ; Vitonis, AF ; Wang-Gohrke, S ; Wentzensen, N ; Whittemore, AS ; Wik, E ; Winterhoff, B ; Woo, YL ; Wu, AH ; Yang, HP ; Zheng, W ; Ziogas, A ; Zulkifli, F ; Goodman, MT ; Hall, P ; Easton, DF ; Pearce, CL ; Berchuck, A ; Chenevix-Trench, G ; Iversen, E ; Monteiro, ANA ; Gayther, SA ; Schildkraut, JM ; Sellers, TA (NATURE PUBLISHING GROUP, 2013-04)
    Genome-wide association studies (GWAS) have identified four susceptibility loci for epithelial ovarian cancer (EOC), with another two suggestive loci reaching near genome-wide significance. We pooled data from a GWAS conducted in North America with another GWAS from the UK. We selected the top 24,551 SNPs for inclusion on the iCOGS custom genotyping array. We performed follow-up genotyping in 18,174 individuals with EOC (cases) and 26,134 controls from 43 studies from the Ovarian Cancer Association Consortium. We validated the two loci at 3q25 and 17q21 that were previously found to have associations close to genome-wide significance and identified three loci newly associated with risk: two loci associated with all EOC subtypes at 8q21 (rs11782652, P = 5.5 × 10(-9)) and 10p12 (rs1243180, P = 1.8 × 10(-8)) and another locus specific to the serous subtype at 17q12 (rs757210, P = 8.1 × 10(-10)). An integrated molecular analysis of genes and regulatory regions at these loci provided evidence for functional mechanisms underlying susceptibility and implicated CHMP4C in the pathogenesis of ovarian cancer.
  • Item
    No Preview Available
    Identification and molecular characterization of a new ovarian cancer susceptibility locus at 17q21.31
    Permuth-Wey, J ; Lawrenson, K ; Shen, HC ; Velkova, A ; Tyrer, JP ; Chen, Z ; Lin, H-Y ; Chen, YA ; Tsai, Y-Y ; Qu, X ; Ramus, SJ ; Karevan, R ; Lee, J ; Lee, N ; Larson, MC ; Aben, KK ; Anton-Culver, H ; Antonenkova, N ; Antoniou, AC ; Armasu, SM ; Bacot, F ; Baglietto, L ; Bandera, EV ; Barnholtz-Sloan, J ; Beckmann, MW ; Birrer, MJ ; Bloom, G ; Bogdanova, N ; Brinton, LA ; Brooks-Wilson, A ; Brown, R ; Butzow, R ; Cai, Q ; Campbell, I ; Chang-Claude, J ; Chanock, S ; Chenevix-Trench, G ; Cheng, JQ ; Cicek, MS ; Coetzee, GA ; Cook, LS ; Couch, FJ ; Cramer, DW ; Cunningham, JM ; Dansonka-Mieszkowska, A ; Despierre, E ; Doherty, JA ; Doerk, T ; du Bois, A ; Duerst, M ; Easton, DF ; Eccles, D ; Edwards, R ; Ekici, AB ; Fasching, PA ; Fenstermacher, DA ; Flanagan, JM ; Garcia-Closas, M ; Gentry-Maharaj, A ; Giles, GG ; Glasspool, RM ; Gonzalez-Bosquet, J ; Goodman, MT ; Gore, M ; Gorski, B ; Gronwald, J ; Hall, P ; Halle, MK ; Harter, P ; Heitz, F ; Hillemanns, P ; Hoatlin, M ; Hogdall, CK ; Hogdall, E ; Hosono, S ; Jakubowska, A ; Jensen, A ; Jim, H ; Kalli, KR ; Karlan, BY ; Kaye, SB ; Kelemen, LE ; Kiemeney, LA ; Kikkawa, F ; Konecny, GE ; Krakstad, C ; Kjaer, SK ; Kupryjanczyk, J ; Lambrechts, D ; Lambrechts, S ; Lancaster, JM ; Le, ND ; Leminen, A ; Levine, DA ; Liang, D ; Lim, BK ; Lin, J ; Lissowska, J ; Lu, KH ; Lubinski, J ; Lurie, G ; Massuger, LFAG ; Matsuo, K ; McGuire, V ; McLaughlin, JR ; Menon, U ; Modugno, F ; Moysich, KB ; Nakanishi, T ; Narod, SA ; Nedergaard, L ; Ness, RB ; Nevanlinna, H ; Nickels, S ; Noushmehr, H ; Odunsi, K ; Olson, SH ; Orlow, I ; Paul, J ; Pearce, CL ; Pejovic, T ; Pelttari, LM ; Pike, MC ; Poole, EM ; Raska, P ; Renner, SP ; Risch, HA ; Rodriguez-Rodriguez, L ; Rossing, MA ; Rudolph, A ; Runnebaum, IB ; Rzepecka, IK ; Salvesen, HB ; Schwaab, I ; Severi, G ; Shridhar, V ; Shu, X-O ; Shvetsov, YB ; Sieh, W ; Song, H ; Southey, MC ; Spiewankiewicz, B ; Stram, D ; Sutphen, R ; Teo, S-H ; Terry, KL ; Tessier, DC ; Thompson, PJ ; Tworoger, SS ; van Altena, AM ; Vergote, I ; Vierkant, RA ; Vincent, D ; Vitonis, AF ; Wang-Gohrke, S ; Weber, RP ; Wentzensen, N ; Whittemore, AS ; Wik, E ; Wilkens, LR ; Winterhoff, B ; Woo, YL ; Wu, AH ; Xiang, Y-B ; Yang, HP ; Zheng, W ; Ziogas, A ; Zulkifli, F ; Phelan, CM ; Iversen, E ; Schildkraut, JM ; Berchuck, A ; Fridley, BL ; Goode, EL ; Pharoah, PDP ; Monteiro, ANA ; Sellers, TA ; Gayther, SA (NATURE RESEARCH, 2013-03)
    Epithelial ovarian cancer (EOC) has a heritable component that remains to be fully characterized. Most identified common susceptibility variants lie in non-protein-coding sequences. We hypothesized that variants in the 3' untranslated region at putative microRNA (miRNA)-binding sites represent functional targets that influence EOC susceptibility. Here, we evaluate the association between 767 miRNA-related single-nucleotide polymorphisms (miRSNPs) and EOC risk in 18,174 EOC cases and 26,134 controls from 43 studies genotyped through the Collaborative Oncological Gene-environment Study. We identify several miRSNPs associated with invasive serous EOC risk (odds ratio=1.12, P=10(-8)) mapping to an inversion polymorphism at 17q21.31. Additional genotyping of non-miRSNPs at 17q21.31 reveals stronger signals outside the inversion (P=10(-10)). Variation at 17q21.31 is associated with neurological diseases, and our collaboration is the first to report an association with EOC susceptibility. An integrated molecular analysis in this region provides evidence for ARHGAP27 and PLEKHM1 as candidate EOC susceptibility genes.
  • Item
    Thumbnail Image
    A simple consensus approach improves somatic mutation prediction accuracy
    Goode, DL ; Hunter, SM ; Doyle, MA ; Ma, T ; Rowley, SM ; Choong, D ; Ryland, GL ; Campbell, IG (BMC, 2013-09-30)
    Differentiating true somatic mutations from artifacts in massively parallel sequencing data is an immense challenge. To develop methods for optimal somatic mutation detection and to identify factors influencing somatic mutation prediction accuracy, we validated predictions from three somatic mutation detection algorithms, MuTect, JointSNVMix2 and SomaticSniper, by Sanger sequencing. Full consensus predictions had a validation rate of >98%, but some partial consensus predictions validated too. In cases of partial consensus, read depth and mapping quality data, along with additional prediction methods, aided in removing inaccurate predictions. Our consensus approach is fast, flexible and provides a high-confidence list of putative somatic mutations.
  • Item
    Thumbnail Image
    Human perforin mutations and susceptibility to multiple primary cancers
    Trapani, JA ; Thia, KYT ; Andrews, M ; Davis, ID ; Gedye, C ; Parente, P ; Svobodova, S ; Chia, J ; Browne, K ; Campbell, IG ; Phillips, WA ; Voskoboinik, I ; Cebon, JS (TAYLOR & FRANCIS INC, 2013-04-01)
    Loss-of-function mutations in the gene coding for perforin (PRF1) markedly reduce the ability of cytotoxic T lymphocytes and natural killer cells to kill target cells, causing immunosuppression and impairing immune regulation. In humans, nearly half of the cases of type 2 familial hemophagocytic lymphohistiocytosis are due to bi-allelic PRF1 mutations. The partial inactivation of PRF1 due to mutations that promote protein misfolding or the common hypomorphic allele coding for the A91V substitution have been associated with lymphoid malignancies in childhood and adolescence. To investigate whether PRF1 mutations also predispose adults to cancer, we genotyped 566 individuals diagnosed with melanoma (101), lymphoma (65), colorectal carcinoma (30) or ovarian cancer (370). The frequency of PRF1 genotypes was similar in all disease groups and 424 matched controls, indicating that the PRF1 status is not associated with an increased susceptibility to these malignancies. However, four out of 15 additional individuals diagnosed with melanoma and B-cell lymphoma during their lifetime expressed either PRF1A91V or the rare pathogenic PRF1R28C variant (p = 0.04), and developed melanoma relatively early in life. Both PRF1A91V- and PRF1R28C-expressing lymphocytes exhibited severely impaired but measurable cytotoxic function. Our results suggest that defects in human PRF1 predispose individuals to develop both melanoma and lymphoma. However, these findings require validation in larger patient cohorts.
  • Item
    No Preview Available
    Functional Analysis of Genes in Regions Commonly Amplified in High-Grade Serous and Endometrioid Ovarian Cancer
    Davis, SJ ; Sheppard, KE ; Pearson, RB ; Campbell, IG ; Gorringe, KL ; Simpson, KJ (AMER ASSOC CANCER RESEARCH, 2013-03-15)
    PURPOSE: Ovarian cancer has the highest mortality rate of all the gynecologic malignancies and is responsible for approximately 140,000 deaths annually worldwide. Copy number amplification is frequently associated with the activation of oncogenic drivers in this tumor type, but their cytogenetic complexity and heterogeneity has made it difficult to determine which gene(s) within an amplicon represent(s) the genuine oncogenic driver. We sought to identify amplicon targets by conducting a comprehensive functional analysis of genes located in the regions of amplification in high-grade serous and endometrioid ovarian tumors. EXPERIMENTAL DESIGN: High-throughput siRNA screening technology was used to systematically assess all genes within regions commonly amplified in high-grade serous and endometrioid cancer. We describe the results from a boutique siRNA screen of 272 genes in a panel of 18 ovarian cell lines. Hits identified by the functional viability screen were further interrogated in primary tumor cohorts to determine the clinical outcomes associated with amplification and gene overexpression. RESULTS: We identified a number of genes as critical for cellular viability when amplified, including URI1, PAK4, GAB2, and DYRK1B. Integration of primary tumor gene expression and outcome data provided further evidence for the therapeutic use of such genes, particularly URI1 and GAB2, which were significantly associated with survival in 2 independent tumor cohorts. CONCLUSION: By taking this integrative approach to target discovery, we have streamlined the translation of high-resolution genomic data into preclinical in vitro studies, resulting in the identification of a number of genes that may be specifically targeted for the treatment of advanced ovarian tumors.
  • Item
    No Preview Available
    Analysis of Over 10,000 Cases Finds No Association between Previously Reported Candidate Polymorphisms and Ovarian Cancer Outcome
    White, KL ; Vierkant, RA ; Fogarty, ZC ; Charbonneau, B ; Block, MS ; Pharoah, PDP ; Chenevix-Trench, G ; Rossing, MA ; Cramer, DW ; Pearce, CL ; Schildkraut, JM ; Menon, U ; Kjaer, SK ; Levine, DA ; Gronwald, J ; Culver, HA ; Whittemore, AS ; Karlan, BY ; Lambrechts, D ; Wentzensen, N ; Kupryjanczyk, J ; Chang-Claude, J ; Bandera, EV ; Hogdall, E ; Heitz, F ; Kaye, SB ; Fasching, PA ; Campbell, I ; Goodman, MT ; Pejovic, T ; Bean, Y ; Lurie, G ; Eccles, D ; Hein, A ; Beckmann, MW ; Ekici, AB ; Paul, J ; Brown, R ; Flanagan, JM ; Harter, P ; Du Bois, A ; Schwaab, I ; Hogdall, CK ; Lundvall, L ; Olson, SH ; Orlow, I ; Paddock, LE ; Rudolph, A ; Eilber, U ; Dansonka-Mieszkowska, A ; Rzepecka, IK ; Ziolkowska-Seta, I ; Brinton, L ; Yang, H ; Garcia-Closas, M ; Despierre, E ; Lambrechts, S ; Vergote, I ; Walsh, C ; Lester, J ; Sieh, W ; McGuire, V ; Rothstein, JH ; Ziogas, A ; Lubinski, J ; Cybulski, C ; Menkiszak, J ; Jensen, A ; Gayther, SA ; Ramus, SJ ; Gentry-Maharaj, A ; Berchuck, A ; Wu, AH ; Pike, MC ; Van denBerg, D ; Terry, KL ; Vitonis, AF ; Doherty, JA ; Johnatty, SE ; Defazio, A ; Song, H ; Tyrer, J ; Sellers, TA ; Phelan, CM ; Kalli, KR ; Cunningham, JM ; Fridley, BL ; Goode, EL (AMER ASSOC CANCER RESEARCH, 2013-05)
    BACKGROUND: Ovarian cancer is a leading cause of cancer-related death among women. In an effort to understand contributors to disease outcome, we evaluated single-nucleotide polymorphisms (SNP) previously associated with ovarian cancer recurrence or survival, specifically in angiogenesis, inflammation, mitosis, and drug disposition genes. METHODS: Twenty-seven SNPs in VHL, HGF, IL18, PRKACB, ABCB1, CYP2C8, ERCC2, and ERCC1 previously associated with ovarian cancer outcome were genotyped in 10,084 invasive cases from 28 studies from the Ovarian Cancer Association Consortium with over 37,000-observed person-years and 4,478 deaths. Cox proportional hazards models were used to examine the association between candidate SNPs and ovarian cancer recurrence or survival with and without adjustment for key covariates. RESULTS: We observed no association between genotype and ovarian cancer recurrence or survival for any of the SNPs examined. CONCLUSIONS: These results refute prior associations between these SNPs and ovarian cancer outcome and underscore the importance of maximally powered genetic association studies. IMPACT: These variants should not be used in prognostic models. Alternate approaches to uncovering inherited prognostic factors, if they exist, are needed.