Sir Peter MacCallum Department of Oncology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 119
  • Item
    Thumbnail Image
    Screening participation for people at increased risk of colorectal cancer due to family history: a systematic review and meta-analysis
    Ouakrim, DA ; Lockett, T ; Boussioutas, A ; Hopper, JL ; Jenkins, MA (SPRINGER, 2013-09)
    We conducted a systematic review and a meta-analysis of observational studies to identify and summarise the level of colorectal cancer (CRC) screening participation for people at increased risk due to family history of the disease. Medline, Cinhal, Embase and PsychInfo databases were comprehensively searched between January 1995 and May 2012 to identify relevant articles. To be included, studies had to report on screening for people who had at least one first-degree relative with CRC and no previous personal diagnosis of the disease. Pooled screening participation levels were calculated for each screening modality. Seventeen studies, accounting for a total of 13,269 subjects with a family history of CRC met the inclusion criteria. Seven studies, including a total of 6,901 subjects had a pooled faecal occult blood testing screening participation (at least once) of 25 % (95 % CI 12-38). Five studies including a total of 5,091 subjects had a pooled sigmoidoscopy-based screening participation (at least once) of 16 % (95 % CI 7-27). Seven studies including a total of 9,965 subjects had pooled participation colonoscopy-based screening (at least once) of 40 % (95 % CI 26-54). There was a significant level of screening heterogeneity between studies. This review identified a substantial underuse of CRC screening for people at increased risk of developing the disease. It highlights the potential opportunity that exists for increasing screening participation among this segment of the population and the need to adjust the current CRC screening policies towards that objective.
  • Item
    Thumbnail Image
    Suboptimal health literacy in patients with lung cancer or head and neck cancer
    Koay, K ; Schofield, P ; Gough, K ; Buchbinder, R ; Rischin, D ; Ball, D ; Corry, J ; Osborne, RH ; Jefford, M (SPRINGER, 2013-08)
    BACKGROUND: Health literacy is the capacity to seek, understand and utilise health information to make informed health decisions. Suboptimal health literacy has been linked to poor health outcomes. This study assessed health literacy in patients treated for head and neck or lung cancer and associations between health literacy and demographic factors and distress levels. METHODS: Consecutive English-speaking patients were approached at Peter MacCallum Cancer Centre. Face-to-face interviews were conducted. Health literacy was assessed using the Shortened Test of Functional Health Literacy in Adults (S-TOFHLA) and Health Literacy Management Scale (HeLMS). Distress was assessed by the Distress Thermometer. RESULTS: Response rate was 73 % (n = 93). Using S-TOFHLA, prevalence of inadequate and marginal health literacy was 5.4 and 6.5 % respectively, and both groups were associated with older age (p = 0.043) and low education level (p = 0.009). Specific assessment of S-TOFHLA revealed that 70 % could not interpret prescription labels. HeLMS reported that 17 % had health literacy difficulties. Low scores on domains of HeLMS were associated with lower education level (p < 0.05) but younger age (p < 0.05). Distress was not associated with S-TOFHLA scores but related to low scores in two domains of HeLMS (p < 0.05). CONCLUSION: Using two different measures, a substantial proportion of patients have poor health literacy abilities and may experience difficulties in accessing health services.
  • Item
    No Preview Available
    Exploration of a Series of 5-Arylidene-2-thioxoimidazolidin-4-ones as Inhibitors of the Cytolytic Protein Perforin
    Spicer, JA ; Lena, G ; Lyons, DM ; Huttunen, KM ; Miller, CK ; O'Connor, PD ; Bull, M ; Helsby, N ; Jamieson, SMF ; Denny, WA ; Ciccone, A ; Browne, KA ; Lopez, JA ; Rudd-Schmidt, J ; Voskoboinik, I ; Trapani, JA (AMER CHEMICAL SOC, 2013-12-12)
    A series of novel 5-arylidene-2-thioxoimidazolidin-4-ones were investigated as inhibitors of the lymphocyte-expressed pore-forming protein perforin. Structure-activity relationships were explored through variation of an isoindolinone or 3,4-dihydroisoquinolinone subunit on a fixed 2-thioxoimidazolidin-4-one/thiophene core. The ability of the resulting compounds to inhibit the lytic activity of both isolated perforin protein and perforin delivered in situ by natural killer cells was determined. A number of compounds showed excellent activity at concentrations that were nontoxic to the killer cells, and several were a significant improvement on previous classes of inhibitors, being substantially more potent and soluble. Representative examples showed rapid and reversible binding to immobilized mouse perforin at low concentrations (≤2.5 μM) by surface plasmon resonance and prevented formation of perforin pores in target cells despite effective target cell engagement, as determined by calcium influx studies. Mouse PK studies of two analogues showed T1/2 values of 1.1-1.2 h (dose of 5 mg/kg i.v.) and MTDs of 60-80 mg/kg (i.p.).
  • Item
    No Preview Available
    DNA methylation in ductal carcinoma in situ of the breast
    Pang, J-MB ; Dobrovic, A ; Fox, SB (BMC, 2013)
    Ductal carcinoma in situ (DCIS) is a non-obligate precursor lesion of invasive carcinoma of the breast. Current prognostic markers based on histopathological examination are unable to accurately predict which DCIS cases will progress to invasive carcinoma or recur after surgical excision. Epigenetic changes have been shown to be a significant driver of tumorigenesis, and DNA methylation of specific gene promoters provides predictive and prognostic markers in many types of cancer, including invasive breast cancer. In general, the spectrum of genes that are methylated in DCIS strongly resembles that seen in invasive ductal carcinoma. The identification of specific prognostic markers in DCIS remains elusive and awaits additional work investigating a large panel of methylatable genes by using sensitive and reproducible technologies. This review critically appraises the role of methylation in DCIS and its use as a biomarker.
  • Item
    No Preview Available
    AKT-independent PI3-K signaling in cancer - emerging role for SGK3.
    Bruhn, MA ; Pearson, RB ; Hannan, RD ; Sheppard, KE (Informa UK Limited, 2013)
    The phosphoinositide 3-kinase (PI3-K) signaling pathway plays an important role in a wide variety of fundamental cellular processes, largely mediated via protein kinase B/v-akt murine thymoma viral oncogene homolog (PKB/AKT) signaling. Given the crucial role of PI3-K/AKT signaling in regulating processes such as cell growth, proliferation, and survival, it is not surprising that components of this pathway are frequently dysregulated in cancer, making the AKT kinase family members important therapeutic targets. The large number of clinical trials currently evaluating PI3-K pathway inhibitors as a therapeutic strategy further emphasizes this. The serum- and glucocorticoid-inducible protein kinase (SGK) family is made up of three isoforms, SGK1, 2, and 3, that are PI3-K-dependent, serine/threonine kinases, with similar substrate specificity to AKT. Consequently, the SGK family also regulates similar cell processes to the AKT kinases, including cell proliferation and survival. Importantly, there is emerging evidence demonstrating that SGK3 plays a critical role in AKT-independent oncogenic signaling. This review will focus on the role of SGK3 as a key effector of AKT-independent PI3-K oncogenic signaling.
  • 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
    Genome-wide association study identifies two susceptibility loci for osteosarcoma
    Savage, SA ; Mirabello, L ; Wang, Z ; Gastier-Foster, JM ; Gorlick, R ; Khanna, C ; Flanagan, AM ; Tirabosco, R ; Andrulis, IL ; Wunder, JS ; Gokgoz, N ; Patino-Garcia, A ; Sierrasesumaga, L ; Lecanda, F ; Kurucu, N ; Ilhan, IE ; Sari, N ; Serra, M ; Hattinger, C ; Picci, P ; Spector, LG ; Barkauskas, DA ; Marina, N ; Caminada de Toledo, SR ; Petrilli, AS ; Amary, MF ; Halai, D ; Thomas, DM ; Douglass, C ; Meltzer, PS ; Jacobs, K ; Chung, CC ; Berndt, SI ; Purdue, MP ; Caporaso, NE ; Tucker, M ; Rothman, N ; Landi, MT ; Silverman, DT ; Kraft, P ; Hunter, DJ ; Malats, N ; Kogevinas, M ; Wacholder, S ; Troisi, R ; Helman, L ; Fraumeni, JF ; Yeager, M ; Hoover, RN ; Chanock, SJ (NATURE PUBLISHING GROUP, 2013-07)
    Osteosarcoma is the most common primary bone malignancy of adolescents and young adults. To better understand the genetic etiology of osteosarcoma, we performed a multistage genome-wide association study consisting of 941 individuals with osteosarcoma (cases) and 3,291 cancer-free adult controls of European ancestry. Two loci achieved genome-wide significance: a locus in the GRM4 gene at 6p21.3 (encoding glutamate receptor metabotropic 4; rs1906953; P = 8.1 × 10⁻⁹) and a locus in the gene desert at 2p25.2 (rs7591996 and rs10208273; P = 1.0 × 10⁻⁸ and 2.9 × 10⁻⁷, respectively). These two loci warrant further exploration to uncover the biological mechanisms underlying susceptibility to osteosarcoma.
  • 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
    Direct repression of MYB by ZEB1 suppresses proliferation and epithelial gene expression during epithelial-to-mesenchymal transition of breast cancer cells
    Hugo, HJ ; Pereira, L ; Suryadinata, R ; Drabsch, Y ; Gonda, TJ ; Gunasinghe, NPAD ; Pinto, C ; Soo, ETL ; van denderen, BJW ; Hill, P ; Ramsay, RG ; Sarcevic, B ; Newgreen, DF ; Thompson, EW (BMC, 2013)
    INTRODUCTION: Epithelial-to-mesenchymal transition (EMT) promotes cell migration and is important in metastasis. Cellular proliferation is often downregulated during EMT, and the reverse transition (MET) in metastases appears to be required for restoration of proliferation in secondary tumors. We studied the interplay between EMT and proliferation control by MYB in breast cancer cells. METHODS: MYB, ZEB1, and CDH1 expression levels were manipulated by lentiviral small-hairpin RNA (shRNA)-mediated knockdown/overexpression, and verified with Western blotting, immunocytochemistry, and qRT-PCR. Proliferation was assessed with bromodeoxyuridine pulse labeling and flow cytometry, and sulforhodamine B assays. EMT was induced with epidermal growth factor for 9 days or by exposure to hypoxia (1% oxygen) for up to 5 days, and assessed with qRT-PCR, cell morphology, and colony morphology. Protein expression in human breast cancers was assessed with immunohistochemistry. ZEB1-MYB promoter binding and repression were determined with Chromatin Immunoprecipitation Assay and a luciferase reporter assay, respectively. Student paired t tests, Mann-Whitney, and repeated measures two-way ANOVA tests determined statistical significance (P < 0.05). RESULTS: Parental PMC42-ET cells displayed higher expression of ZEB1 and lower expression of MYB than did the PMC42-LA epithelial variant. Knockdown of ZEB1 in PMC42-ET and MDA-MB-231 cells caused increased expression of MYB and a transition to a more epithelial phenotype, which in PMC42-ET cells was coupled with increased proliferation. Indeed, we observed an inverse relation between MYB and ZEB1 expression in two in vitro EMT cell models, in matched human breast tumors and lymph node metastases, and in human breast cancer cell lines. Knockdown of MYB in PMC42-LA cells (MYBsh-LA) led to morphologic changes and protein expression consistent with an EMT. ZEB1 expression was raised in MYBsh-LA cells and significantly repressed in MYB-overexpressing MDA-MB-231 cells, which also showed reduced random migration and a shift from mesenchymal to epithelial colony morphology in two dimensional monolayer cultures. Finally, we detected binding of ZEB1 to MYB promoter in PMC42-ET cells, and ZEB1 overexpression repressed MYB promoter activity. CONCLUSIONS: This work identifies ZEB1 as a transcriptional repressor of MYB and suggests a reciprocal MYB-ZEB1 repressive relation, providing a mechanism through which proliferation and the epithelial phenotype may be coordinately modulated in breast cancer cells.