Sir Peter MacCallum Department of Oncology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 36
  • Item
    Thumbnail Image
    Going to extremes: determinants of extraordinary response and survival in patients with cancer
    Saner, FAM ; Herschtal, A ; Nelson, BH ; deFazio, A ; Goode, EL ; Ramus, SJ ; Pandey, A ; Beach, JA ; Fereday, S ; Berchuck, A ; Lheureux, S ; Pearce, CL ; Pharoah, PD ; Pike, MC ; Garsed, DW ; Bowtell, DDL (NATURE PUBLISHING GROUP, 2019-06)
    Research into factors affecting treatment response or survival in patients with cancer frequently involves cohorts that span the most common range of clinical outcomes, as such patients are most readily available for study. However, attention has turned to highly unusual patients who have exceptionally favourable or atypically poor responses to treatment and/or overall survival, with the expectation that patients at the extremes may provide insights that could ultimately improve the outcome of individuals with more typical disease trajectories. While clinicians can often recount surprising patients whose clinical journey was very unusual, given known clinical characteristics and prognostic indicators, there is a lack of consensus among researchers on how best to define exceptional patients, and little has been proposed for the optimal design of studies to identify factors that dictate unusual outcome. In this Opinion article, we review different approaches to identifying exceptional patients with cancer and possible study designs to investigate extraordinary clinical outcomes. We discuss pitfalls with finding these rare patients, including challenges associated with accrual of patients across different treatment centres and time periods. We describe recent molecular and immunological factors that have been identified as contributing to unusual patient outcome and make recommendations for future studies on these intriguing patients.
  • Item
    Thumbnail Image
    Critical questions in ovarian cancer research and treatment: Report of an American Association for Cancer Research Special Conference
    Bast, RC ; Matulonis, UA ; Sood, AK ; Ahmed, AA ; Amobi, AE ; Balkwill, FR ; Wielgos-Bonvallet, M ; Bowtell, DDL ; Brenton, JD ; Brugge, JS ; Coleman, RL ; Draetta, GF ; Doberstein, K ; Drapkin, RI ; Eckert, MA ; Edwards, RP ; Elias, KM ; Ennis, D ; Futreal, A ; Gershenson, DM ; Greenberg, RA ; Huntsman, DG ; Ji, JXY ; Kohn, EC ; Iavarone, C ; Lengyel, ER ; Levine, DA ; Lord, CJ ; Lu, Z ; Mills, GB ; Modugno, F ; Nelson, BH ; Odunsi, K ; Pilsworth, JA ; Rottapel, RK ; Powell, DJ ; Shen, L ; Shih, I-M ; Spriggs, DR ; Walton, J ; Zhang, K ; Zhang, R ; Zou, L (WILEY, 2019-06-15)
    Substantial progress has been made in understanding ovarian cancer at the molecular and cellular level. Significant improvement in 5-year survival has been achieved through cytoreductive surgery, combination platinum-based chemotherapy, and more effective treatment of recurrent cancer, and there are now more than 280,000 ovarian cancer survivors in the United States. Despite these advances, long-term survival in late-stage disease has improved little over the last 4 decades. Poor outcomes relate, in part, to late stage at initial diagnosis, intrinsic drug resistance, and the persistence of dormant drug-resistant cancer cells after primary surgery and chemotherapy. Our ability to accelerate progress in the clinic will depend on the ability to answer several critical questions regarding this disease. To assess current answers, an American Association for Cancer Research Special Conference on "Critical Questions in Ovarian Cancer Research and Treatment" was held in Pittsburgh, Pennsylvania, on October 1-3, 2017. Although clinical, translational, and basic investigators conducted much of the discussion, advocates participated in the meeting, and many presentations were directly relevant to patient care, including treatment with poly adenosine diphosphate ribose polymerase (PARP) inhibitors, attempts to improve immunotherapy by overcoming the immune suppressive effects of the microenvironment, and a better understanding of the heterogeneity of the disease.
  • Item
    No Preview Available
    BRCA1 Promoter Methylation and Clinical Outcomes in Ovarian Cancer: An Individual Patient Data Meta-Analysis
    Kalachand, RD ; Stordal, B ; Madden, S ; Chandler, B ; Cunningham, J ; Goode, EL ; Ruscito, I ; Braicu, E ; Sehouli, J ; Ignatov, A ; Yu, H ; Katsaros, D ; Mills, GB ; Lu, KH ; Carey, MS ; Timms, KM ; Kupryjanczyk, J ; Rzepecka, IK ; Podgorska, A ; McAlpine, JN ; Swisher, EM ; Bernards, SS ; O'Riain, C ; O'Toole, S ; O'Leary, JJ ; Bowtell, DD ; Thomas, DM ; Prieske, K ; Joosse, SA ; Woelber, L ; Chaudhry, P ; Hafner, N ; Runnebaum, IB ; Hennessy, BT (OXFORD UNIV PRESS INC, 2020-12)
    BACKGROUND: BRCA1 methylation has been associated with homologous recombination deficiency, a biomarker of platinum sensitivity. Studies evaluating BRCA1-methylated tubal and ovarian cancer (OC) do not consistently support improved survival following platinum chemotherapy. We examine the characteristics of BRCA1-methylated OC in a meta-analysis of individual participant data. METHODS: Data of 2636 participants across 15 studies were analyzed. BRCA1-methylated tumors were defined according to their original study. Associations between BRCA1 methylation and clinicopathological characteristics were evaluated. The effects of methylation on overall survival (OS) and progression-free survival (PFS) were examined using mixed-effects models. All statistical tests were 2-sided. RESULTS: 430 (16.3%) tumors were BRCA1-methylated. BRCA1 methylation was associated with younger age and advanced-stage, high-grade serous OC. There were no survival differences between BRCA1-methylated and non-BRCA1-methylated OC (median PFS = 20.0 vs 18.5 months, hazard ratio [HR] = 1.01, 95% CI = 0.87 to 1.16; P = .98; median OS = 46.6 vs 48.0 months, HR = 1.02, 95% CI = 0.87 to 1.18; P = .96). Where BRCA1/2 mutations were evaluated (n = 1248), BRCA1 methylation displayed no survival advantage over BRCA1/2-intact (BRCA1/2 wild-type non-BRCA1-methylated) OC. Studies used different methods to define BRCA1 methylation. Where BRCA1 methylation was determined using methylation-specific polymerase chain reaction and gel electrophoresis (n = 834), it was associated with improved survival (PFS: HR = 0.80, 95% CI = 0.66 to 0.97; P = .02; OS: HR = 0.80, 95% CI = 0.63 to 1.00; P = .05) on mixed-effects modeling. CONCLUSION: BRCA1-methylated OC displays similar clinicopathological features to BRCA1-mutated OC but is not associated with survival. Heterogeneity within BRCA1 methylation assays influences associations. Refining these assays may better identify cases with silenced BRCA1 function and improved patient outcomes.
  • Item
    Thumbnail Image
    Signal Transduction Pathway Activity in High-Grade, Serous Ovarian Carcinoma Reveals a More Favorable Prognosis in Tumors with Low PI3K and High NF-κB Pathway Activity: A Novel Approach to a Long-Standing Enigma
    van Lieshout, L ; van de Stolpe, A ; van der Ploeg, P ; Bowtell, D ; de Hullu, J ; Piek, J (MDPI, 2020-09)
    We investigated signal transduction pathway (STP) activity in high-grade serous ovarian carcinoma (HGSC) in relation to progression-free survival (PFS) and overall survival (OS). We made use of signal transduction pathway activity analysis (STA analysis), a novel method to quantify functional STP activity. Activity of the following pathways was measured: androgen receptor (AR), estrogen receptor (ER), phosphoinositide 3-kinase (PI3K), Hedgehog (Hh), Notch, nuclear factor-kappa B (NF-κB), transforming growth factor beta (TGF-β), and Wnt. We selected HGSC samples from publicly available datasets of ovarian cancer tissue, and used repeated k-means clustering to identify pathway activity clusters. PFS and OS of the clusters were analyzed. We used a subset of publicly available dataset GSE9891 (n = 140), where repeated k-means clustering based on PI3K and NF-κB pathway activity in HGSC samples resulted in two stable clusters. The cluster with low PI3K and high NF-κB pathway activity (n = 72) had a more favorable prognosis for both PFS (p = 0.004) and OS (p = 0.001) compared to the high-PI3K and low-NF-κB pathway activity cluster (n = 68). The low PI3K and high NF-κB pathway activity of the favorable prognosis cluster may indicate a more active immune response, while the high PI3K and low NF-κB pathway activity of the unfavorable prognosis cluster may indicate high cell division.
  • Item
    Thumbnail Image
    ABCC4/MRP4 contributes to the aggressiveness of Myc-associated epithelial ovarian cancer
    Jung, M ; Gao, J ; Cheung, L ; Bongers, A ; Somers, K ; Clifton, M ; Ramsay, EE ; Russell, AJ ; Valli, E ; Gifford, AJ ; George, J ; Kennedy, CJ ; Wakefield, MJ ; Topp, M ; Ho, G-Y ; Scott, CL ; Bowtell, DD ; deFazio, A ; Norris, MD ; Haber, M ; Henderson, MJ (WILEY, 2020-10-15)
    Epithelial ovarian cancer (EOC) is a complex disease comprising discrete histological and molecular subtypes, for which survival rates remain unacceptably low. Tailored approaches for this deadly heterogeneous disease are urgently needed. Efflux pumps belonging to the ATP-binding cassette (ABC) family of transporters are known for roles in both drug resistance and cancer biology and are also highly targetable. Here we have investigated the association of ABCC4/MRP4 expression to clinical outcome and its biological function in endometrioid and serous tumors, common histological subtypes of EOC. We found high expression of ABCC4/MRP4, previously shown to be directly regulated by c-Myc/N-Myc, was associated with poor prognosis in endometrioid EOC (P = .001) as well as in a subset of serous EOC with a "high-MYCN" profile (C5/proliferative; P = .019). Transient siRNA-mediated suppression of MRP4 in EOC cells led to reduced growth, migration and invasion, with the effects being most pronounced in endometrioid and C5-like serous cells compared to non-C5 serous EOC cells. Sustained knockdown of MRP4 also sensitized endometrioid cells to MRP4 substrate drugs. Furthermore, suppression of MRP4 decreased the growth of patient-derived EOC cells in vivo. Together, our findings provide the first evidence that MRP4 plays an important role in the biology of Myc-associated ovarian tumors and highlight this transporter as a potential therapeutic target for EOC.
  • Item
    Thumbnail Image
    Therapeutic options for mucinous ovarian carcinoma
    Gorringe, KL ; Cheasley, D ; Wakefield, MJ ; Ryland, GL ; Allan, PE ; Alsop, K ; Amarasinghe, KC ; Ananda, S ; Bowtell, DDL ; Christie, M ; Chiew, Y-E ; Churchman, M ; DeFazio, A ; Fereday, S ; Gilks, CB ; Gourley, C ; Hadley, AM ; Hendley, J ; Hunter, SM ; Kaufmann, SH ; Kennedy, CJ ; Kobel, M ; Le Page, C ; Li, J ; Lupat, R ; McNally, OM ; McAlpine, JN ; Pyman, J ; Rowley, SM ; Salazar, C ; Saunders, H ; Semple, T ; Stephens, AN ; Thio, N ; Torres, MC ; Traficante, N ; Zethoven, M ; Antill, YC ; Campbell, IG ; Scott, CL (ACADEMIC PRESS INC ELSEVIER SCIENCE, 2020-03)
    OBJECTIVE: Mucinous ovarian carcinoma (MOC) is an uncommon ovarian cancer histotype that responds poorly to conventional chemotherapy regimens. Although long overall survival outcomes can occur with early detection and optimal surgical resection, recurrent and advanced disease are associated with extremely poor survival. There are no current guidelines specifically for the systemic management of recurrent MOC. We analyzed data from a large cohort of women with MOC to evaluate the potential for clinical utility from a range of systemic agents. METHODS: We analyzed gene copy number (n = 191) and DNA sequencing data (n = 184) from primary MOC to evaluate signatures of mismatch repair deficiency and homologous recombination deficiency, and other genetic events. Immunohistochemistry data were collated for ER, CK7, CK20, CDX2, HER2, PAX8 and p16 (n = 117-166). RESULTS: Molecular aberrations noted in MOC that suggest a match with current targeted therapies include amplification of ERBB2 (26.7%) and BRAF mutation (9%). Observed genetic events that suggest potential efficacy for agents currently in clinical trials include: KRAS/NRAS mutations (66%), TP53 missense mutation (49%), RNF43 mutation (11%), ARID1A mutation (10%), and PIK3CA/PTEN mutation (9%). Therapies exploiting homologous recombination deficiency (HRD) may not be effective in MOC, as only 1/191 had a high HRD score. Mismatch repair deficiency was similarly rare (1/184). CONCLUSIONS: Although genetically diverse, MOC has several potential therapeutic targets. Importantly, the lack of response to platinum-based therapy observed clinically corresponds to the lack of a genomic signature associated with HRD, and MOC are thus also unlikely to respond to PARP inhibition.
  • Item
    Thumbnail Image
    Association of p16 expression with prognosis varies across ovarian carcinoma histotypes: an Ovarian Tumor Tissue Analysis consortium study
    Rambau, PF ; Vierkant, RA ; Intermaggio, MP ; Kelemen, LE ; Goodman, MT ; Herpel, E ; Pharoah, PD ; Kommoss, S ; Jimenez-Linan, M ; Karlan, BY ; Gentry-Maharaj, A ; Menon, U ; Polo, SH ; Candido dos Reis, FJ ; Doherty, JA ; Gayther, SA ; Sharma, R ; Larson, MC ; Harnett, PR ; Hatfield, E ; de Andrade, JM ; Nelson, GS ; Steed, H ; Schildkraut, JM ; Carney, ME ; Hogdall, E ; Whittemore, AS ; Widschwendter, M ; Kennedy, CJ ; Wang, F ; Wang, Q ; Wang, C ; Armasu, SM ; Daley, F ; Coulson, P ; Jones, ME ; Anglesio, MS ; Chow, C ; de Fazio, A ; Garcia-Closas, M ; Brucker, SY ; Cybulski, C ; Harris, HR ; Hartkopf, AD ; Huzarski, T ; Jensen, A ; Lubinski, J ; Oszurek, O ; Benitez, J ; Mina, F ; Staebler, A ; Taran, FA ; Pasternak, J ; Talhouk, A ; Rossing, MA ; Hendley, J ; Edwards, RP ; Fereday, S ; Modugno, F ; Ness, RB ; Sieh, W ; El-Bahrawy, MA ; Winham, SJ ; Lester, J ; Kjaer, SK ; Gronwald, J ; Sinn, P ; Fasching, PA ; Chang-Claude, J ; Moysich, KB ; Bowtell, DD ; Hernandez, BY ; Luk, H ; Behrens, S ; Shah, M ; Jung, A ; Ghatage, P ; Alsop, J ; Alsop, K ; Garcia-Donas, J ; Thompson, PJ ; Swerdlow, AJ ; Karpinskyj, C ; Cazorla-Jimenez, A ; Garcia, MJ ; Deen, S ; Wilkens, LR ; Palacios, J ; Berchuck, A ; Koziak, JM ; Brenton, JD ; Cook, LS ; Goode, EL ; Huntsman, DG ; Ramus, SJ ; Koebel, M (WILEY, 2018-10)
    We aimed to validate the prognostic association of p16 expression in ovarian high-grade serous carcinomas (HGSC) and to explore it in other ovarian carcinoma histotypes. p16 protein expression was assessed by clinical-grade immunohistochemistry in 6525 ovarian carcinomas including 4334 HGSC using tissue microarrays from 24 studies participating in the Ovarian Tumor Tissue Analysis consortium. p16 expression patterns were interpreted as abnormal (either overexpression referred to as block expression or absence) or normal (heterogeneous). CDKN2A (which encodes p16) mRNA expression was also analyzed in a subset (n = 2280) mostly representing HGSC (n = 2010). Association of p16 expression with overall survival (OS) was determined within histotypes as was CDKN2A expression for HGSC only. p16 block expression was most frequent in HGSC (56%) but neither protein nor mRNA expression was associated with OS. However, relative to heterogeneous expression, block expression was associated with shorter OS in endometriosis-associated carcinomas, clear cell [hazard ratio (HR): 2.02, 95% confidence (CI) 1.47-2.77, p < 0.001] and endometrioid (HR: 1.88, 95% CI 1.30-2.75, p = 0.004), while absence was associated with shorter OS in low-grade serous carcinomas (HR: 2.95, 95% CI 1.61-5.38, p = 0.001). Absence was most frequent in mucinous carcinoma (50%), and was not associated with OS in this histotype. The prognostic value of p16 expression is histotype-specific and pattern dependent. We provide definitive evidence against an association of p16 expression with survival in ovarian HGSC as previously suggested. Block expression of p16 in clear cell and endometrioid carcinoma should be further validated as a prognostic marker, and absence in low-grade serous carcinoma justifies CDK4 inhibition.
  • Item
    Thumbnail Image
    Genomic Analysis Using Regularized Regression in High-Grade Serous Ovarian Cancer
    Natanzon, Y ; Earp, M ; Cunningham, JM ; Kalli, KR ; Wang, C ; Armasu, SM ; Larson, MC ; Bowtell, DDL ; Garsed, DW ; Fridley, BL ; Winham, SJ ; Goode, EL (SAGE PUBLICATIONS LTD, 2018-02-01)
    High-grade serous ovarian cancer (HGSOC) is a complex disease in which initiation and progression have been associated with copy number alterations, epigenetic processes, and, to a lesser extent, germline variation. We hypothesized that, when summarized at the gene level, tumor methylation and germline genetic variation, alone or in combination, influence tumor gene expression in HGSOC. We used Elastic Net (ENET) penalized regression method to evaluate these associations and adjust for somatic copy number in 3 independent data sets comprising tumors from more than 470 patients. Penalized regression models of germline variation, with or without methylation, did not reveal a role in HGSOC gene expression. However, we observed significant association between regional methylation and expression of 5 genes (WDPCP, KRT6C, BRCA2, EFCAB13, and ZNF283). CpGs retained in ENET model for BRCA2 and ZNF283 appeared enriched in several regulatory elements, suggesting that regularized regression may provide a novel utility for integrative genomic analysis.
  • Item
    Thumbnail Image
    MyD88 and TLR4 Expression in Epithelial Ovarian Cancer
    Block, MS ; Vierkant, RA ; Rambau, PF ; Winham, SJ ; Wagner, P ; Traficante, N ; Toloczko, A ; Tiezzi, DG ; Taran, FA ; Sinn, P ; Sieh, W ; Sharma, R ; Rothstein, JH ; Ramon y Cajal, T ; Paz-Ares, L ; Oszurek, O ; Orsulic, S ; Ness, RB ; Nelson, G ; Modugno, F ; Menkiszak, J ; McGuire, V ; McCauley, BM ; Mack, M ; Lubinski, J ; Longacre, TA ; Li, Z ; Lester, J ; Kennedy, CJ ; Kalli, KR ; Jung, AY ; Johnatty, SE ; Jimenez-Linan, M ; Jensen, A ; Intermaggio, MP ; Hung, J ; Herpel, E ; Hernandez, BY ; Hartkopf, AD ; Harnett, PR ; Ghatage, P ; Garcia-Bueno, JM ; Gao, B ; Fereday, S ; Eilber, U ; Edwards, RP ; de Sousa, CB ; de Andrade, JM ; Chudecka-Glaz, A ; Chenevix-Trench, G ; Cazorla, A ; Brucker, SY ; Alsop, J ; Whittemore, AS ; Steed, H ; Staebler, A ; Moysich, KB ; Menon, U ; Koziak, JM ; Kommoss, S ; Kjaer, SK ; Kelemen, LE ; Karlan, BY ; Huntsman, DG ; Hogdall, E ; Gronwald, J ; Goodman, MT ; Gilks, B ; Jose Garcia, M ; Fasching, PA ; de Fazio, A ; Deen, S ; Chang-Claude, J ; dos Reis, FJC ; Campbell, IG ; Brenton, JD ; Bowtell, DD ; Benitez, J ; Pharoah, PDP ; Kobel, M ; Ramus, SJ ; Goode, EL (ELSEVIER SCIENCE INC, 2018-03)
    OBJECTIVE: To evaluate myeloid differentiation primary response gene 88 (MyD88) and Toll-like receptor 4 (TLR4) expression in relation to clinical features of epithelial ovarian cancer, histologic subtypes, and overall survival. PATIENTS AND METHODS: We conducted centralized immunohistochemical staining, semi-quantitative scoring, and survival analysis in 5263 patients participating in the Ovarian Tumor Tissue Analysis consortium. Patients were diagnosed between January 1, 1978, and December 31, 2014, including 2865 high-grade serous ovarian carcinomas (HGSOCs), with more than 12,000 person-years of follow-up time. Tissue microarrays were stained for MyD88 and TLR4, and staining intensity was classified using a 2-tiered system for each marker (weak vs strong). RESULTS: Expression of MyD88 and TLR4 was similar in all histotypes except clear cell ovarian cancer, which showed reduced expression compared with other histotypes (P<.001 for both). In HGSOC, strong MyD88 expression was modestly associated with shortened overall survival (hazard ratio [HR], 1.13; 95% CI, 1.01-1.26; P=.04) but was also associated with advanced stage (P<.001). The expression of TLR4 was not associated with survival. In low-grade serous ovarian cancer (LGSOC), strong expression of both MyD88 and TLR4 was associated with favorable survival (HR [95% CI], 0.49 [0.29-0.84] and 0.44 [0.21-0.89], respectively; P=.009 and P=.02, respectively). CONCLUSION: Results are consistent with an association between strong MyD88 staining and advanced stage and poorer survival in HGSOC and demonstrate correlation between strong MyD88 and TLR4 staining and improved survival in LGSOC, highlighting the biological differences between the 2 serous histotypes.
  • Item
    Thumbnail Image
    Chemotherapy weakly contributes to predicted neoantigen expression in ovarian cancer
    O'Donnell, T ; Christie, EL ; Ahuja, A ; Buros, J ; Aksoy, BA ; Bowtell, DDL ; Snyder, A ; Hammerbacher, J (BMC, 2018-01-22)
    BACKGROUND: Patients with highly mutated tumors, such as melanoma or smoking-related lung cancer, have higher rates of response to immune checkpoint blockade therapy, perhaps due to increased neoantigen expression. Many chemotherapies including platinum compounds are known to be mutagenic, but the impact of standard treatment protocols on mutational burden and resulting neoantigen expression in most human cancers is unknown. METHODS: We sought to quantify the effect of chemotherapy treatment on computationally predicted neoantigen expression for high grade serous ovarian carcinoma patients enrolled in the Australian Ovarian Cancer Study. In this series, 35 of 114 samples were collected after exposure to chemotherapy; 14 are matched with an untreated sample from the same patient. Our approach integrates whole genome and RNA sequencing of bulk tumor samples with class I MHC binding prediction and mutational signatures extracted from studies of chemotherapy-exposed Caenorhabditis elegans and Gallus gallus cells. We additionally investigated the relationship between neoantigens, tumor infiltrating immune cells estimated from RNA-seq with CIBERSORT, and patient survival. RESULTS: Greater neoantigen burden and CD8+ T cell infiltration in primary, pre-treatment samples were independently associated with improved survival. Relapse samples collected after chemotherapy harbored a median of 78% more expressed neoantigens than untreated primary samples, a figure that combines the effects of chemotherapy and other processes operative during relapse. The contribution from chemotherapy-associated signatures was small, accounting for a mean of 5% (range 0-16) of the expressed neoantigen burden in relapse samples. In both treated and untreated samples, most neoantigens were attributed to COSMIC Signature (3), associated with BRCA disruption, Signature (1), associated with a slow mutagenic process active in healthy tissue, and Signature (8), of unknown etiology. CONCLUSION: Relapsed ovarian cancers harbor more predicted neoantigens than primary tumors, but the increase is due to pre-existing mutational processes, not mutagenesis from chemotherapy.