Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Kathryn Alsop is active.

Publication


Featured researches published by Kathryn Alsop.


Journal of Clinical Oncology | 2012

BRCA Mutation Frequency and Patterns of Treatment Response in BRCA Mutation–Positive Women With Ovarian Cancer: A Report From the Australian Ovarian Cancer Study Group

Kathryn Alsop; Sian Fereday; Cliff Meldrum; Anna deFazio; Catherine Emmanuel; Joshy George; Alexander Dobrovic; Michael J. Birrer; Penelope M. Webb; Colin J.R. Stewart; Michael Friedlander; Stephen B. Fox; David Bowtell; Gillian Mitchell

PURPOSE The frequency of BRCA1 and BRCA2 germ-line mutations in women with ovarian cancer is unclear; reports vary from 3% to 27%. The impact of germ-line mutation on response requires further investigation to understand its impact on treatment planning and clinical trial design. PATIENTS AND METHODS Women with nonmucinous ovarian carcinoma (n = 1,001) enrolled onto a population-based, case-control study were screened for point mutations and large deletions in both genes. Survival outcomes and responses to multiple lines of chemotherapy were assessed. RESULTS Germ-line mutations were found in 14.1% of patients overall, including 16.6% of serous cancer patients (high-gradeserous, 17.1%); [corrected] 44% had no reported family history of breast orovarian cancer.Patients carrying germ-line mutations had improved rates of progression-free and overall survival. In the relapse setting, patients carrying mutations more frequently responded to both platin- and nonplatin-based regimens than mutation-negative patients, even in patients with early relapse after primary treatment. Mutation-negative patients who responded to multiple cycles of platin-based treatment were more likely to carry somatic BRCA1/2 mutations. CONCLUSION BRCA mutation status has a major influence on survival in ovarian cancer patients and should be an additional stratification factor in clinical trials. Treatment outcomes in BRCA1/2 carriers challenge conventional definitions of platin resistance, and mutation status may be able to contribute to decision making and systemic therapy selection in the relapse setting. Our data, together with the advent of poly(ADP-ribose) polymerase inhibitor trials, supports the recommendation that germ-line BRCA1/2 testing should be offered to all women diagnosed with nonmucinous, ovarian carcinoma, regardless of family history.


Nature | 2015

Whole–genome characterization of chemoresistant ovarian cancer

Ann-Marie Patch; Elizabeth L. Christie; Dariush Etemadmoghadam; Dale W. Garsed; Joshy George; Sian Fereday; Katia Nones; Prue Cowin; Kathryn Alsop; Peter Bailey; Karin S. Kassahn; Felicity Newell; Michael Quinn; Stephen Kazakoff; Kelly Quek; Charlotte Wilhelm-Benartzi; Ed Curry; Huei San Leong; Anne Hamilton; Linda Mileshkin; George Au-Yeung; Catherine Kennedy; Jillian Hung; Yoke-Eng Chiew; Paul Harnett; Michael Friedlander; Jan Pyman; Stephen M. Cordner; Patricia O’Brien; Jodie Leditschke

Patients with high-grade serous ovarian cancer (HGSC) have experienced little improvement in overall survival, and standard treatment has not advanced beyond platinum-based combination chemotherapy, during the past 30 years. To understand the drivers of clinical phenotypes better, here we use whole-genome sequencing of tumour and germline DNA samples from 92 patients with primary refractory, resistant, sensitive and matched acquired resistant disease. We show that gene breakage commonly inactivates the tumour suppressors RB1, NF1, RAD51B and PTEN in HGSC, and contributes to acquired chemotherapy resistance. CCNE1 amplification was common in primary resistant and refractory disease. We observed several molecular events associated with acquired resistance, including multiple independent reversions of germline BRCA1 or BRCA2 mutations in individual patients, loss of BRCA1 promoter methylation, an alteration in molecular subtype, and recurrent promoter fusion associated with overexpression of the drug efflux pump MDR1.


Clinical Cancer Research | 2009

Integrated Genome-Wide DNA Copy Number and Expression Analysis Identifies Distinct Mechanisms of Primary Chemoresistance in Ovarian Carcinomas

Dariush Etemadmoghadam; Anna deFazio; Rameen Beroukhim; Craig H. Mermel; Joshy George; Gad Getz; Richard W. Tothill; Aikou Okamoto; Maria B. Ræder; Paul Harnett; Stephen Lade; Lars A. Akslen; Anna V. Tinker; Bianca Locandro; Kathryn Alsop; Yoke-Eng Chiew; Nadia Traficante; Sian Fereday; Daryl S. Johnson; Stephen B. Fox; William R. Sellers; Mitsuyoshi Urashima; Helga B. Salvesen; Matthew Meyerson; David Bowtell

Purpose: A significant number of women with serous ovarian cancer are intrinsically refractory to platinum-based treatment. We analyzed somatic DNA copy number variation and gene expression data to identify key mechanisms associated with primary resistance in advanced-stage serous cancers. Experimental Design: Genome-wide copy number variation was measured in 118 ovarian tumors using high-resolution oligonucleotide microarrays. A well-defined subset of 85 advanced-stage serous tumors was then used to relate copy number variation to primary resistance to treatment. The discovery-based approach was complemented by quantitative-PCR copy number analysis of 12 candidate genes as independent validation of previously reported associations with clinical outcome. Likely copy number variation targets and tumor molecular subtypes were further characterized by gene expression profiling. Results: Amplification of 19q12, containing cyclin E (CCNE1), and 20q11.22-q13.12, mapping immediately adjacent to the steroid receptor coactivator NCOA3, was significantly associated with poor response to primary treatment. Other genes previously associated with copy number variation and clinical outcome in ovarian cancer were not associated with primary treatment resistance. Chemoresistant tumors with high CCNE1 copy number and protein expression were associated with increased cellular proliferation but so too was a subset of treatment-responsive patients, suggesting a cell-cycle independent role for CCNE1 in modulating chemoresponse. Patients with a poor clinical outcome without CCNE1 amplification overexpressed genes involved in extracellular matrix deposition. Conclusions: We have identified two distinct mechanisms of primary treatment failure in serous ovarian cancer, involving CCNE1 amplification and enhanced extracellular matrix deposition. CCNE1 copy number is validated as a dominant marker of patient outcome in ovarian cancer.


Journal of the National Cancer Institute | 2015

Germline Mutations in the BRIP1, BARD1, PALB2, and NBN Genes in Women With Ovarian Cancer

Susan J. Ramus; Honglin Song; Ed Dicks; Jonathan Tyrer; Adam N. Rosenthal; Maria P. Intermaggio; Lindsay Fraser; Aleksandra Gentry-Maharaj; Jane Hayward; Susan Philpott; Christopher E. Anderson; Christopher K. Edlund; David V. Conti; Patricia Harrington; Daniel Barrowdale; David Bowtell; Kathryn Alsop; Gillian Mitchell; Mine S. Cicek; Julie M. Cunningham; Brooke L. Fridley; Jennifer Alsop; Mercedes Jimenez-Linan; Samantha Poblete; S.B. Lele; Lara E. Sucheston-Campbell; Kirsten B. Moysich; Weiva Sieh; Valerie McGuire; Jenny Lester

BACKGROUND Epithelial ovarian cancer (EOC) is the most lethal gynecological malignancy, responsible for 13 000 deaths per year in the United States. Risk prediction based on identifying germline mutations in ovarian cancer susceptibility genes could have a clinically significant impact on reducing disease mortality. METHODS Next generation sequencing was used to identify germline mutations in the coding regions of four candidate susceptibility genes-BRIP1, BARD1, PALB2 and NBN-in 3236 invasive EOC case patients and 3431 control patients of European origin, and in 2000 unaffected high-risk women from a clinical screening trial of ovarian cancer (UKFOCSS). For each gene, we estimated the prevalence and EOC risks and evaluated associations between germline variant status and clinical and epidemiological risk factor information. All statistical tests were two-sided. RESULTS We found an increased frequency of deleterious mutations in BRIP1 in case patients (0.9%) and in the UKFOCSS participants (0.6%) compared with control patients (0.09%) (P = 1 x 10(-4) and 8 x 10(-4), respectively), but no differences for BARD1 (P = .39), NBN1 ( P = .61), or PALB2 (P = .08). There was also a difference in the frequency of rare missense variants in BRIP1 between case patients and control patients (P = 5.5 x 10(-4)). The relative risks associated with BRIP1 mutations were 11.22 for invasive EOC (95% confidence interval [CI] = 3.22 to 34.10, P = 1 x 10(-4)) and 14.09 for high-grade serous disease (95% CI = 4.04 to 45.02, P = 2 x 10(-5)). Segregation analysis in families estimated the average relative risks in BRIP1 mutation carriers compared with the general population to be 3.41 (95% CI = 2.12 to 5.54, P = 7×10(-7)). CONCLUSIONS Deleterious germline mutations in BRIP1 are associated with a moderate increase in EOC risk. These data have clinical implications for risk prediction and prevention approaches for ovarian cancer and emphasize the critical need for risk estimates based on very large sample sizes before genes of moderate penetrance have clinical utility in cancer prevention.


Journal of Clinical Oncology | 2015

Contribution of Germline Mutations in the RAD51B, RAD51C, and RAD51D Genes to Ovarian Cancer in the Population

Honglin Song; Ed Dicks; Susan J. Ramus; Jonathan Tyrer; Maria P. Intermaggio; Jane Hayward; Christopher K. Edlund; David V. Conti; Patricia Harrington; Lindsay Fraser; Susan Philpott; Christopher N. G. Anderson; Adam Rosenthal; Aleksandra Gentry-Maharaj; David Bowtell; Kathryn Alsop; Mine S. Cicek; Julie M. Cunningham; Brooke L. Fridley; Jennifer Alsop; Mercedes Jimenez-Linan; Estrid Høgdall; C Hogdall; Allan Jensen; Susanne Kriiger Kjaer; Jan Lubinski; Tomasz Huzarski; Anna Jakubowska; Jacek Gronwald; Samantha Poblete

PURPOSE The aim of this study was to estimate the contribution of deleterious mutations in the RAD51B, RAD51C, and RAD51D genes to invasive epithelial ovarian cancer (EOC) in the population and in a screening trial of individuals at high risk of ovarian cancer. PATIENTS AND METHODS The coding sequence and splice site boundaries of the three RAD51 genes were sequenced and analyzed in germline DNA from a case-control study of 3,429 patients with invasive EOC and 2,772 controls as well as in 2,000 unaffected women who were BRCA1/BRCA2 negative from the United Kingdom Familial Ovarian Cancer Screening Study (UK_FOCSS) after quality-control analysis. RESULTS In the case-control study, we identified predicted deleterious mutations in 28 EOC cases (0.82%) compared with three controls (0.11%; P < .001). Mutations in EOC cases were more frequent in RAD51C (14 occurrences, 0.41%) and RAD51D (12 occurrences, 0.35%) than in RAD51B (two occurrences, 0.06%). RAD51C mutations were associated with an odds ratio of 5.2 (95% CI, 1.1 to 24; P = .035), and RAD51D mutations conferred an odds ratio of 12 (95% CI, 1.5 to 90; P = .019). We identified 13 RAD51 mutations (0.65%) in unaffected UK_FOCSS participants (RAD51C, n = 7; RAD51D, n = 5; and RAD51B, n = 1), which was a significantly greater rate than in controls (P < .001); furthermore, RAD51 mutation carriers were more likely than noncarriers to have a family history of ovarian cancer (P < .001). CONCLUSION These results confirm that RAD51C and RAD51D are moderate ovarian cancer susceptibility genes and suggest that they confer levels of risk of EOC that may warrant their use alongside BRCA1 and BRCA2 in routine clinical genetic testing.


Clinical Cancer Research | 2012

Profiles of Genomic Instability in High-Grade Serous Ovarian Cancer Predict Treatment Outcome

Zhigang C. Wang; Nicolai Juul Birkbak; Aedín C. Culhane; Ronny Drapkin; Aquila Fatima; Ruiyang Tian; Matthew Schwede; Kathryn Alsop; Kathryn E. Daniels; Huiying Piao; Joy Liu; Dariush Etemadmoghadam; Alexander Miron; Helga B. Salvesen; Gillian Mitchell; Anna deFazio; John Quackenbush; Ross S. Berkowitz; James Dirk Iglehart; David Bowtell; Ursula A. Matulonis

Purpose: High-grade serous cancer (HGSC) is the most common cancer of the ovary and is characterized by chromosomal instability. Defects in homologous recombination repair (HRR) are associated with genomic instability in HGSC, and are exploited by therapy targeting DNA repair. Defective HRR causes uniparental deletions and loss of heterozygosity (LOH). Our purpose is to profile LOH in HGSC and correlate our findings to clinical outcome, and compare HGSC and high-grade breast cancers. Experimental Design: We examined LOH and copy number changes using single nucleotide polymorphism array data from three HGSC cohorts and compared results to a cohort of high-grade breast cancers. The LOH profiles in HGSC were matched to chemotherapy resistance and progression-free survival (PFS). Results: LOH-based clustering divided HGSC into two clusters. The major group displayed extensive LOH and was further divided into two subgroups. The second group contained remarkably less LOH. BRCA1 promoter methylation was associated with the major group. LOH clusters were reproducible when validated in two independent HGSC datasets. LOH burden in the major cluster of HGSC was similar to triple-negative, and distinct from other high-grade breast cancers. Our analysis revealed an LOH cluster with lower treatment resistance and a significant correlation between LOH burden and PFS. Conclusions: Separating HGSC by LOH-based clustering produces remarkably stable subgroups in three different cohorts. Patients in the various LOH clusters differed with respect to chemotherapy resistance, and the extent of LOH correlated with PFS. LOH burden may indicate vulnerability to treatment targeting DNA repair, such as PARP1 inhibitors. Clin Cancer Res; 18(20); 5806–15. ©2012 AACR.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Synthetic lethality between CCNE1 amplification and loss of BRCA1

Dariush Etemadmoghadam; Barbara A. Weir; G. Au-Yeung; Kathryn Alsop; Gillian Mitchell; Joshy George; Sally J. Davis; Alan D. D'Andrea; Kaylene J. Simpson; William C. Hahn; David Bowtell

Significance Women with high-grade serous ovarian cancer (HGSC) harboring Cyclin E1 (CCNE1) gene amplification generally face a poor clinical outcome. These tumors comprise a significant group of ∼20% of HGSCs that are not associated with BRCA1/2 mutation and are unlikely to respond to standard cytotoxic or poly-ADP-ribose polymerase inhibitors. We identified a specific dependency on BRCA1 and members of the ubiquitin pathway in CCNE1-amplified tumors. The requirement for BRCA1 seems to account for the mutual exclusivity of mutations observed in primary tumors. We propose a unique therapeutic strategy involving inhibition of the proteasome and homologous recombination function with bortezomib. Our findings are likely to have relevance to the treatment of other tumor types with CCNE1 amplification, including triple negative breast cancer. High-grade serous ovarian cancers (HGSCs) are characterized by a high frequency of TP53 mutations, BRCA1/2 inactivation, homologous recombination dysfunction, and widespread copy number changes. Cyclin E1 (CCNE1) gene amplification has been reported to occur independently of BRCA1/2 mutation, and it is associated with primary treatment failure and reduced patient survival. Insensitivity of CCNE1-amplified tumors to platinum cross-linking agents may be partly because of an intact BRCA1/2 pathway. Both BRCA1/2 dysfunction and CCNE1 amplification are known to promote genomic instability and tumor progression. These events may be mutually exclusive, because either change provides a path to tumor development, with no selective advantage to having both mutations. Using data from a genome-wide shRNA synthetic lethal screen, we show that BRCA1 and members of the ubiquitin pathway are selectively required in cancers that harbor CCNE1 amplification. Furthermore, we show specific sensitivity of CCNE1-amplified tumor cells to the proteasome inhibitor bortezomib. These findings provide an explanation for the observed mutual exclusivity of CCNE1 amplification and BRCA1/2 loss in HGSC and suggest a unique therapeutic approach for treatment-resistant CCNE1-amplified tumors.


The Journal of Pathology | 2012

Tandem duplication of chromosomal segments is common in ovarian and breast cancer genomes

David J. McBride; Dariush Etemadmoghadam; Susanna L. Cooke; Kathryn Alsop; Joshy George; Adam Butler; Juok Cho; Danushka Galappaththige; Christopher Greenman; Karen Howarth; King Wai Lau; Charlotte K.Y. Ng; Keiran Raine; Jon Teague; David C. Wedge; Xavier Caubit; Michael R. Stratton; James D. Brenton; Peter J. Campbell; P. Andrew Futreal; David Bowtell

The application of paired‐end next generation sequencing approaches has made it possible to systematically characterize rearrangements of the cancer genome to base‐pair level. Utilizing this approach, we report the first detailed analysis of ovarian cancer rearrangements, comparing high‐grade serous and clear cell cancers, and these histotypes with other solid cancers. Somatic rearrangements were systematically characterized in eight high‐grade serous and five clear cell ovarian cancer genomes and we report here the identification of > 600 somatic rearrangements. Recurrent rearrangements of the transcriptional regulator gene, TSHZ3, were found in three of eight serous cases. Comparison to breast, pancreatic and prostate cancer genomes revealed that a subset of ovarian cancers share a marked tandem duplication phenotype with triple‐negative breast cancers. The tandem duplication phenotype was not linked to BRCA1/2 mutation, suggesting that other common mechanisms or carcinogenic exposures are operative. High‐grade serous cancers arising in women with germline BRCA1 or BRCA2 mutation showed a high frequency of small chromosomal deletions. These findings indicate that BRCA1/2 germline mutation may contribute to widespread structural change and that other undefined mechanism(s), which are potentially shared with triple‐negative breast cancer, promote tandem chromosomal duplications that sculpt the ovarian cancer genome. Copyright


Clinical Cancer Research | 2015

Germline Mutation in BRCA1 or BRCA2 and Ten-Year Survival for Women Diagnosed with Epithelial Ovarian Cancer

Francisco José Candido dos Reis; Honglin Song; Ellen L. Goode; Julie M. Cunningham; Brooke L. Fridley; Melissa C. Larson; Kathryn Alsop; Ed Dicks; Patricia Harrington; Susan J. Ramus; Anna de Fazio; Gillian Mitchell; Sian Fereday; Kelly L. Bolton; Charlie Gourley; Caroline O. Michie; Beth Y. Karlan; Jenny Lester; C. Walsh; Ilana Cass; Håkan Olsson; Martin Gore; Javier Benitez; María J. García; Irene L. Andrulis; Anna Marie Mulligan; Gord Glendon; Ignacio Blanco; Conxi Lázaro; Alice S. Whittemore

Purpose: To analyze the effect of germline mutations in BRCA1 and BRCA2 on mortality in patients with ovarian cancer up to 10 years after diagnosis. Experimental Design: We used unpublished survival time data for 2,242 patients from two case–control studies and extended survival time data for 4,314 patients from previously reported studies. All participants had been screened for deleterious germline mutations in BRCA1 and BRCA2. Survival time was analyzed for the combined data using Cox proportional hazard models with BRCA1 and BRCA2 as time-varying covariates. Competing risks were analyzed using Fine and Gray model. Results: The combined 10-year overall survival rate was 30% [95% confidence interval (CI), 28%–31%] for non-carriers, 25% (95% CI, 22%–28%) for BRCA1 carriers, and 35% (95% CI, 30%–41%) for BRCA2 carriers. The HR for BRCA1 was 0.53 at time zero and increased over time becoming greater than one at 4.8 years. For BRCA2, the HR was 0.42 at time zero and increased over time (predicted to become greater than 1 at 10.5 years). The results were similar when restricted to 3,202 patients with high-grade serous tumors and to ovarian cancer–specific mortality. Conclusions: BRCA1/2 mutations are associated with better short-term survival, but this advantage decreases over time and in BRCA1 carriers is eventually reversed. This may have important implications for therapy of both primary and relapsed disease and for analysis of long-term survival in clinical trials of new agents, particularly those that are effective in BRCA1/2 mutation carriers. Clin Cancer Res; 21(3); 652–7. ©2014 AACR.


Molecular Oncology | 2014

Molecular correlates of platinum response in human high-grade serous ovarian cancer patient-derived xenografts

Monique Topp; Lynne Hartley; Michele Cook; Valerie Heong; Emma Boehm; Lauren McShane; Jan Pyman; Orla McNally; Sumitra Ananda; Marisol Harrell; Dariush Etemadmoghadam; Laura Galletta; Kathryn Alsop; Gillian Mitchell; Stephen B. Fox; J. B. Kerr; Karla J. Hutt; Scott H. Kaufmann; Elizabeth M. Swisher; David Bowtell; Matthew J. Wakefield; Clare L. Scott

Improvement in the ability to target underlying drivers and vulnerabilities of high‐grade serous ovarian cancer (HG‐SOC) requires the development of molecularly annotated pre‐clinical models reflective of clinical responses.

Collaboration


Dive into the Kathryn Alsop's collaboration.

Top Co-Authors

Avatar

David Bowtell

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Gillian Mitchell

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Sian Fereday

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joshy George

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Stephen B. Fox

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nadia Traficante

Peter MacCallum Cancer Centre

View shared research outputs
Researchain Logo
Decentralizing Knowledge