Network


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

Hotspot


Dive into the research topics where Ann-Marie Patch is active.

Publication


Featured researches published by Ann-Marie Patch.


Nature | 2015

Whole genomes redefine the mutational landscape of pancreatic cancer

Nicola Waddell; Marina Pajic; Ann-Marie Patch; David K. Chang; Karin S. Kassahn; Peter Bailey; Amber L. Johns; David Miller; Katia Nones; Kelly Quek; Michael Quinn; Alan Robertson; Muhammad Z.H. Fadlullah; Timothy J. C. Bruxner; Angelika N. Christ; Ivon Harliwong; Senel Idrisoglu; Suzanne Manning; Craig Nourse; Ehsan Nourbakhsh; Shivangi Wani; Peter J. Wilson; Emma Markham; Nicole Cloonan; Matthew J. Anderson; J. Lynn Fink; Oliver Holmes; Stephen Kazakoff; Conrad Leonard; Felicity Newell

Pancreatic cancer remains one of the most lethal of malignancies and a major health burden. We performed whole-genome sequencing and copy number variation (CNV) analysis of 100 pancreatic ductal adenocarcinomas (PDACs). Chromosomal rearrangements leading to gene disruption were prevalent, affecting genes known to be important in pancreatic cancer (TP53, SMAD4, CDKN2A, ARID1A and ROBO2) and new candidate drivers of pancreatic carcinogenesis (KDM6A and PREX2). Patterns of structural variation (variation in chromosomal structure) classified PDACs into 4 subtypes with potential clinical utility: the subtypes were termed stable, locally rearranged, scattered and unstable. A significant proportion harboured focal amplifications, many of which contained druggable oncogenes (ERBB2, MET, FGFR1, CDK6, PIK3R3 and PIK3CA), but at low individual patient prevalence. Genomic instability co-segregated with inactivation of DNA maintenance genes (BRCA1, BRCA2 or PALB2) and a mutational signature of DNA damage repair deficiency. Of 8 patients who received platinum therapy, 4 of 5 individuals with these measures of defective DNA maintenance responded.


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.


Nature Communications | 2014

Genomic catastrophes frequently arise in esophageal adenocarcinoma and drive tumorigenesis

Katia Nones; Nicola Waddell; Nicci Wayte; Ann-Marie Patch; Peter Bailey; Felicity Newell; Oliver Holmes; J. Lynn Fink; Michael Quinn; Yue Hang Tang; Guy Lampe; Kelly Quek; Kelly A. Loffler; Suzanne Manning; Senel Idrisoglu; David Miller; Qinying Xu; Nick Waddell; Peter Wilson; Timothy J. C. Bruxner; Angelika N. Christ; Ivon Harliwong; Craig Nourse; Ehsan Nourbakhsh; Matthew Anderson; Stephen Kazakoff; Conrad Leonard; Scott Wood; Peter T. Simpson; Lynne Reid

Oesophageal adenocarcinoma (EAC) incidence is rapidly increasing in Western countries. A better understanding of EAC underpins efforts to improve early detection and treatment outcomes. While large EAC exome sequencing efforts to date have found recurrent loss-of-function mutations, oncogenic driving events have been underrepresented. Here we use a combination of whole-genome sequencing (WGS) and single-nucleotide polymorphism-array profiling to show that genomic catastrophes are frequent in EAC, with almost a third (32%, n = 40/123) undergoing chromothriptic events. WGS of 22 EAC cases show that catastrophes may lead to oncogene amplification through chromothripsis-derived double-minute chromosome formation (MYC and MDM2) or breakage-fusion-bridge (KRAS, MDM2 and RFC3). Telomere shortening is more prominent in EACs bearing localized complex rearrangements. Mutational signature analysis also confirms that extreme genomic instability in EAC can be driven by somatic BRCA2 mutations. These findings suggest that genomic catastrophes have a significant role in the malignant transformation of EAC.


Genome Medicine | 2013

Clinical and molecular characterization of HER2 amplified-pancreatic cancer

Angela Chou; Nicola Waddell; Mark J. Cowley; Anthony J. Gill; David K. Chang; Ann-Marie Patch; Katia Nones; Jianmin Wu; Mark Pinese; Amber L. Johns; David Miller; Karin S. Kassahn; Adnan Nagrial; Harpreet Wasan; David Goldstein; Christopher W. Toon; Venessa T. Chin; Lorraine A. Chantrill; Jeremy L. Humphris; R. Scott Mead; Ilse Rooman; Jaswinder S. Samra; Marina Pajic; Elizabeth A. Musgrove; John V. Pearson; Adrienne Morey; Sean M. Grimmond; Andrew V. Biankin

BackgroundPancreatic cancer is one of the most lethal and molecularly diverse malignancies. Repurposing of therapeutics that target specific molecular mechanisms in different disease types offers potential for rapid improvements in outcome. Although HER2 amplification occurs in pancreatic cancer, it is inadequately characterized to exploit the potential of anti-HER2 therapies.MethodsHER2 amplification was detected and further analyzed using multiple genomic sequencing approaches. Standardized reference laboratory assays defined HER2 amplification in a large cohort of patients (n = 469) with pancreatic ductal adenocarcinoma (PDAC).ResultsAn amplified inversion event (1 MB) was identified at the HER2 locus in a patient with PDAC. Using standardized laboratory assays, we established diagnostic criteria for HER2 amplification in PDAC, and observed a prevalence of 2%. Clinically, HER2- amplified PDAC was characterized by a lack of liver metastases, and a preponderance of lung and brain metastases. Excluding breast and gastric cancer, the incidence of HER2-amplified cancers in the USA is >22,000 per annum.ConclusionsHER2 amplification occurs in 2% of PDAC, and has distinct features with implications for clinical practice. The molecular heterogeneity of PDAC implies that even an incidence of 2% represents an attractive target for anti-HER2 therapies, as options for PDAC are limited. Recruiting patients based on HER2 amplification, rather than organ of origin, could make trials of anti-HER2 therapies feasible in less common cancer types.


Gastroenterology | 2017

Hypermutation In Pancreatic Cancer

Jeremy L. Humphris; Ann-Marie Patch; Katia Nones; Peter Bailey; Amber L. Johns; Skye McKay; David K. Chang; David Miller; Marina Pajic; Karin S. Kassahn; Michael Quinn; Timothy J. C. Bruxner; Angelika N. Christ; Ivon Harliwong; Senel Idrisoglu; Suzanne Manning; Craig Nourse; Ehsan Nourbakhsh; Andrew Stone; Peter J. Wilson; Matthew Anderson; J. Lynn Fink; Oliver Holmes; Stephen Kazakoff; Conrad Leonard; Felicity Newell; Nick Waddell; Scott Wood; Ronald S. Mead; Qinying Xu

Pancreatic cancer is molecularly diverse, with few effective therapies. Increased mutation burden and defective DNA repair are associated with response to immune checkpoint inhibitors in several other cancer types. We interrogated 385 pancreatic cancer genomes to define hypermutation and its causes. Mutational signatures inferring defects in DNA repair were enriched in those with the highest mutation burdens. Mismatch repair deficiency was identified in 1% of tumors harboring different mechanisms of somatic inactivation of MLH1 and MSH2. Defining mutation load in individual pancreatic cancers and the optimal assay for patient selection may inform clinical trial design for immunotherapy in pancreatic cancer.


The Journal of Pathology | 2015

Integrated genomic and transcriptomic analysis of human brain metastases identifies alterations of potential clinical significance

Jodi M. Saunus; Michael Quinn; Ann-Marie Patch; John V. Pearson; Peter Bailey; Katia Nones; Amy E. McCart Reed; David Miller; Peter Wilson; Fares Al-Ejeh; Mythily Mariasegaram; Queenie Lau; Teresa Withers; Rosalind L. Jeffree; Lynne Reid; Leonard Da Silva; Admire Matsika; Colleen Niland; Margaret C. Cummings; Timothy J. C. Bruxner; Angelika N. Christ; Ivon Harliwong; Senel Idrisoglu; Suzanne Manning; Craig Nourse; Ehsan Nourbakhsh; Shivangi Wani; Matthew J. Anderson; J. Lynn Fink; Oliver Holmes

Treatment options for patients with brain metastases (BMs) have limited efficacy and the mortality rate is virtually 100%. Targeted therapy is critically under‐utilized, and our understanding of mechanisms underpinning metastatic outgrowth in the brain is limited. To address these deficiencies, we investigated the genomic and transcriptomic landscapes of 36 BMs from breast, lung, melanoma and oesophageal cancers, using DNA copy‐number analysis and exome‐ and RNA‐sequencing. The key findings were as follows. (a) Identification of novel candidates with possible roles in BM development, including the significantly mutated genes DSC2, ST7, PIK3R1 and SMC5, and the DNA repair, ERBB–HER signalling, axon guidance and protein kinase‐A signalling pathways. (b) Mutational signature analysis was applied to successfully identify the primary cancer type for two BMs with unknown origins. (c) Actionable genomic alterations were identified in 31/36 BMs (86%); in one case we retrospectively identified ERBB2 amplification representing apparent HER2 status conversion, then confirmed progressive enrichment for HER2‐positivity across four consecutive metastatic deposits by IHC and SISH, resulting in the deployment of HER2‐targeted therapy for the patient. (d) In the ERBB/HER pathway, ERBB2 expression correlated with ERBB3 (r2 = 0.496; p < 0.0001) and HER3 and HER4 were frequently activated in an independent cohort of 167 archival BM from seven primary cancer types: 57.6% and 52.6% of cases were phospho‐HER3Y1222 or phospho‐HER4Y1162 membrane‐positive, respectively. The HER3 ligands NRG1/2 were barely detectable by RNAseq, with NRG1 (8p12) genomic loss in 63.6% breast cancer‐BMs, suggesting a microenvironmental source of ligand. In summary, this is the first study to characterize the genomic landscapes of BM. The data revealed novel candidates, potential clinical applications for genomic profiling of resectable BMs, and highlighted the possibility of therapeutically targeting HER3, which is broadly over‐expressed and activated in BMs, independent of primary site and systemic therapy. Copyright


PLOS ONE | 2013

Somatic Point Mutation Calling in Low Cellularity Tumors

Karin S. Kassahn; Oliver Holmes; Katia Nones; Ann-Marie Patch; David Miller; Angelika N. Christ; Ivon Harliwong; Timothy J. C. Bruxner; Qinying Xu; Matthew Anderson; Scott Wood; Conrad Leonard; Darrin Taylor; Felicity Newell; Sarah Song; Senel Idrisoglu; Craig Nourse; Ehsan Nourbakhsh; Suzanne Manning; Shivangi Wani; Anita L Steptoe; Marina Pajic; Mark J. Cowley; Mark Pinese; David K. Chang; Anthony J. Gill; Amber L. Johns; Jianmin Wu; Peter Wilson; Lynn Fink

Somatic mutation calling from next-generation sequencing data remains a challenge due to the difficulties of distinguishing true somatic events from artifacts arising from PCR, sequencing errors or mis-mapping. Tumor cellularity or purity, sub-clonality and copy number changes also confound the identification of true somatic events against a background of germline variants. We have developed a heuristic strategy and software (http://www.qcmg.org/bioinformatics/qsnp/) for somatic mutation calling in samples with low tumor content and we show the superior sensitivity and precision of our approach using a previously sequenced cell line, a series of tumor/normal admixtures, and 3,253 putative somatic SNVs verified on an orthogonal platform.


Genome Medicine | 2017

Lost in translation: returning germline genetic results in genome-scale cancer research.

Amber L. Johns; Skye McKay; Jeremy L. Humphris; Mark Pinese; Lorraine A. Chantrill; R. Scott Mead; Katherine L. Tucker; Lesley Andrews; Annabel Goodwin; Conrad Leonard; Hilda High; Katia Nones; Ann-Marie Patch; Neil D. Merrett; Nick Pavlakis; Karin S. Kassahn; Jaswinder S. Samra; David Miller; David K. Chang; Marina Pajic; John V. Pearson; Sean M. Grimmond; Nicola Waddell; Nikolajs Zeps; Anthony J. Gill; Andrew V. Biankin

BackgroundThe return of research results (RoR) remains a complex and well-debated issue. Despite the debate, actual data related to the experience of giving individual results back, and the impact these results may have on clinical care and health outcomes, is sorely lacking. Through the work of the Australian Pancreatic Cancer Genome Initiative (APGI) we: (1) delineate the pathway back to the patient where actionable research data were identified; and (2) report the clinical utilisation of individual results returned. Using this experience, we discuss barriers and opportunities associated with a comprehensive process of RoR in large-scale genomic research that may be useful for others developing their own policies.MethodsWe performed whole-genome (n = 184) and exome (n = 208) sequencing of matched tumour-normal DNA pairs from 392 patients with sporadic pancreatic cancer (PC) as part of the APGI. We identified pathogenic germline mutations in candidate genes (n = 130) with established predisposition to PC or medium–high penetrance genes with well-defined cancer associated syndromes or phenotypes. Variants from candidate genes were annotated and classified according to international guidelines. Variants were considered actionable if clinical utility was established, with regard to prevention, diagnosis, prognostication and/or therapy.ResultsA total of 48,904 germline variants were identified, with 2356 unique variants undergoing annotation and in silico classification. Twenty cases were deemed actionable and were returned via previously described RoR framework, representing an actionable finding rate of 5.1%. Overall, 1.78% of our cohort experienced clinical benefit from RoR.ConclusionReturning research results within the context of large-scale genomics research is a labour-intensive, highly variable, complex operation. Results that warrant action are not infrequent, but the prevalence of those who experience a clinical difference as a result of returning individual results is currently low.


Clinical Cancer Research | 2017

Homologous Recombination DNA Repair Pathway Disruption and Retinoblastoma Protein Loss Are Associated with Exceptional Survival in High-Grade Serous Ovarian Cancer.

Dale W. Garsed; Kathryn Alsop; Sian Fereday; Catherine Emmanuel; Catherine J. Kennedy; Dariush Etemadmoghadam; Bo Gao; Val Gebski; Valérie Garès; Elizabeth L. Christie; Maartje C.A. Wouters; Katy Milne; Joshy George; Ann-Marie Patch; Jason Li; Gisela Mir Arnau; Timothy Semple; Sreeja R. Gadipally; Yoke-Eng Chiew; Joy Hendley; Thomas Mikeska; Giada V. Zapparoli; Kaushalya C. Amarasinghe; Sean M. Grimmond; John V. Pearson; Nicola Waddell; Jillian Hung; Colin J.R. Stewart; Raghwa Sharma; Prue E. Allan

Purpose: Women with epithelial ovarian cancer generally have a poor prognosis; however, a subset of patients has an unexpected dramatic and durable response to treatment. We sought to identify clinical, pathological, and molecular determinants of exceptional survival in women with high-grade serous cancer (HGSC), a disease associated with the majority of ovarian cancer deaths. Experimental Design: We evaluated the histories of 2,283 ovarian cancer patients and, after applying stringent clinical and pathological selection criteria, identified 96 with HGSC that represented significant outliers in terms of treatment response and overall survival. Patient samples were characterized immunohistochemically and by genome sequencing. Results: Different patterns of clinical response were seen: long progression-free survival (Long-PFS), multiple objective responses to chemotherapy (Multiple Responder), and/or greater than 10-year overall survival (Long-Term Survivors). Pathogenic germline and somatic mutations in genes involved in homologous recombination (HR) repair were enriched in all three groups relative to a population-based series. However, 29% of 10-year survivors lacked an identifiable HR pathway alteration, and tumors from these patients had increased Ki-67 staining. CD8+ tumor-infiltrating lymphocytes were more commonly present in Long-Term Survivors. RB1 loss was associated with long progression-free and overall survival. HR deficiency and RB1 loss were correlated, and co-occurrence was significantly associated with prolonged survival. Conclusions: There was diversity in the clinical trajectory of exceptional survivors associated with multiple molecular determinants of exceptional outcome in HGSC patients. Concurrent HR deficiency and RB1 loss were associated with favorable outcomes, suggesting that co-occurrence of specific mutations might mediate durable responses in such patients. Clin Cancer Res; 24(3); 569–80. ©2017 AACR. See related commentary by Peng and Mills, p. 508


American Journal of Human Genetics | 2017

Long Noncoding RNAs CUPID1 and CUPID2 Mediate Breast Cancer Risk at 11q13 by Modulating the Response to DNA Damage

Joshua A. Betts; Mahdi Moradi Marjaneh; Fares Al-Ejeh; Yi Chieh Lim; Wei Shi; Haran Sivakumaran; Romain Tropée; Ann-Marie Patch; Michael B. Clark; Nenad Bartonicek; Adrian P. Wiegmans; Kristine M. Hillman; Susanne Kaufmann; Amanda L. Bain; Brian S. Gloss; Joanna Crawford; Stephen Kazakoff; Shivangi Wani; Shu Wen Wen; Bryan W. Day; Andreas Möller; Nicole Cloonan; John V. Pearson; Melissa A. Brown; Timothy R. Mercer; Nicola Waddell; Kum Kum Khanna; Eloise Dray; Marcel E. Dinger; Stacey L. Edwards

Breast cancer risk is strongly associated with an intergenic region on 11q13. We have previously shown that the strongest risk-associated SNPs fall within a distal enhancer that regulates CCND1. Here, we report that, in addition to regulating CCND1, this enhancer regulates two estrogen-regulated long noncoding RNAs, CUPID1 and CUPID2. We provide evidence that the risk-associated SNPs are associated with reduced chromatin looping between the enhancer and the CUPID1 and CUPID2 bidirectional promoter. We further show that CUPID1 and CUPID2 are predominantly expressed in hormone-receptor-positive breast tumors and play a role in modulating pathway choice for the repair of double-strand breaks. These data reveal a mechanism for the involvement of this region in breast cancer.

Collaboration


Dive into the Ann-Marie Patch's collaboration.

Top Co-Authors

Avatar

Katia Nones

University of Queensland

View shared research outputs
Top Co-Authors

Avatar

Nicola Waddell

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

John V. Pearson

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Oliver Holmes

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Stephen Kazakoff

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David Miller

University of Queensland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Craig Nourse

University of Queensland

View shared research outputs
Researchain Logo
Decentralizing Knowledge