Network


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

Hotspot


Dive into the research topics where Nicola Waddell is active.

Publication


Featured researches published by Nicola Waddell.


Nature | 2013

Signatures of mutational processes in human cancer

Ludmil B. Alexandrov; Serena Nik-Zainal; David C. Wedge; Samuel Aparicio; Sam Behjati; Andrew V. Biankin; Graham R. Bignell; Niccolo Bolli; Åke Borg; Anne Lise Børresen-Dale; Sandrine Boyault; Birgit Burkhardt; Adam Butler; Carlos Caldas; Helen Davies; Christine Desmedt; Roland Eils; Jórunn Erla Eyfjörd; John A. Foekens; Mel Greaves; Fumie Hosoda; Barbara Hutter; Tomislav Ilicic; Sandrine Imbeaud; Marcin Imielinsk; Natalie Jäger; David T. W. Jones; David Jones; Stian Knappskog; Marcel Kool

All cancers are caused by somatic mutations; however, understanding of the biological processes generating these mutations is limited. The catalogue of somatic mutations from a cancer genome bears the signatures of the mutational processes that have been operative. Here we analysed 4,938,362 mutations from 7,042 cancers and extracted more than 20 distinct mutational signatures. Some are present in many cancer types, notably a signature attributed to the APOBEC family of cytidine deaminases, whereas others are confined to a single cancer class. Certain signatures are associated with age of the patient at cancer diagnosis, known mutagenic exposures or defects in DNA maintenance, but many are of cryptic origin. In addition to these genome-wide mutational signatures, hypermutation localized to small genomic regions, ‘kataegis’, is found in many cancer types. The results reveal the diversity of mutational processes underlying the development of cancer, with potential implications for understanding of cancer aetiology, prevention and therapy.


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.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Sleeping Beauty mutagenesis reveals cooperating mutations and pathways in pancreatic adenocarcinoma

Karen M. Mann; Jerrold M. Ward; Christopher Chin Kuan Yew; Anne N. Kovochich; David W. Dawson; Michael A. Black; Benjamin T. Brett; Todd Sheetz; Adam J. Dupuy; David K. Chang; Andrew V. Biankin; Nicola Waddell; Karin S. Kassahn; Sean M. Grimmond; Alistair G. Rust; David J. Adams; Nancy A. Jenkins; Neal G. Copeland

Pancreatic cancer is one of the most deadly cancers affecting the Western world. Because the disease is highly metastatic and difficult to diagnosis until late stages, the 5-y survival rate is around 5%. The identification of molecular cancer drivers is critical for furthering our understanding of the disease and development of improved diagnostic tools and therapeutics. We have conducted a mutagenic screen using Sleeping Beauty (SB) in mice to identify new candidate cancer genes in pancreatic cancer. By combining SB with an oncogenic Kras allele, we observed highly metastatic pancreatic adenocarcinomas. Using two independent statistical methods to identify loci commonly mutated by SB in these tumors, we identified 681 loci that comprise 543 candidate cancer genes (CCGs); 75 of these CCGs, including Mll3 and Ptk2, have known mutations in human pancreatic cancer. We identified point mutations in human pancreatic patient samples for another 11 CCGs, including Acvr2a and Map2k4. Importantly, 10% of the CCGs are involved in chromatin remodeling, including Arid4b, Kdm6a, and Nsd3, and all SB tumors have at least one mutated gene involved in this process; 20 CCGs, including Ctnnd1, Fbxo11, and Vgll4, are also significantly associated with poor patient survival. SB mutagenesis provides a rich resource of mutations in potential cancer drivers for cross-comparative analyses with ongoing sequencing efforts in human pancreatic adenocarcinoma.


Nature | 2017

Whole-genome landscapes of major melanoma subtypes

Nicholas K. Hayward; James S. Wilmott; Nicola Waddell; Peter A. Johansson; Matthew A. Field; Katia Nones; Ann Marie Patch; Hojabr Kakavand; Ludmil B. Alexandrov; Hazel Burke; Valerie Jakrot; Stephen Kazakoff; Oliver Holmes; Conrad Leonard; Radhakrishnan Sabarinathan; Loris Mularoni; Scott Wood; Qinying Xu; Nick Waddell; Varsha Tembe; Gulietta M. Pupo; Ricardo De Paoli-Iseppi; Ricardo E. Vilain; Ping Shang; Loretta Lau; Rebecca A. Dagg; Sarah-Jane Schramm; Antonia L. Pritchard; Ken Dutton-Regester; Felicity Newell

Melanoma of the skin is a common cancer only in Europeans, whereas it arises in internal body surfaces (mucosal sites) and on the hands and feet (acral sites) in people throughout the world. Here we report analysis of whole-genome sequences from cutaneous, acral and mucosal subtypes of melanoma. The heavily mutated landscape of coding and non-coding mutations in cutaneous melanoma resolved novel signatures of mutagenesis attributable to ultraviolet radiation. However, acral and mucosal melanomas were dominated by structural changes and mutation signatures of unknown aetiology, not previously identified in melanoma. The number of genes affected by recurrent mutations disrupting non-coding sequences was similar to that affected by recurrent mutations to coding sequences. Significantly mutated genes included BRAF, CDKN2A, NRAS and TP53 in cutaneous melanoma, BRAF, NRAS and NF1 in acral melanoma and SF3B1 in mucosal melanoma. Mutations affecting the TERT promoter were the most frequent of all; however, neither they nor ATRX mutations, which correlate with alternative telomere lengthening, were associated with greater telomere length. Most melanomas had potentially actionable mutations, most in components of the mitogen-activated protein kinase and phosphoinositol kinase pathways. The whole-genome mutation landscape of melanoma reveals diverse carcinogenic processes across its subtypes, some unrelated to sun exposure, and extends potential involvement of the non-coding genome in its pathogenesis.


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.


Nature Communications | 2015

A comprehensive assessment of somatic mutation detection in cancer using whole-genome sequencing

Tyler Alioto; Ivo Buchhalter; Sophia Derdak; Barbara Hutter; Matthew Eldridge; Eivind Hovig; Lawrence E. Heisler; Timothy Beck; Jared T. Simpson; Laurie Tonon; Anne Sophie Sertier; Ann Marie Patch; Natalie Jäger; Philip Ginsbach; Ruben M. Drews; Nagarajan Paramasivam; Rolf Kabbe; Sasithorn Chotewutmontri; Nicolle Diessl; Christopher Previti; Sabine Schmidt; Benedikt Brors; Lars Feuerbach; Michael Heinold; Susanne Gröbner; Andrey Korshunov; Patrick Tarpey; Adam Butler; Jonathan Hinton; David Jones

As whole-genome sequencing for cancer genome analysis becomes a clinical tool, a full understanding of the variables affecting sequencing analysis output is required. Here using tumour-normal sample pairs from two different types of cancer, chronic lymphocytic leukaemia and medulloblastoma, we conduct a benchmarking exercise within the context of the International Cancer Genome Consortium. We compare sequencing methods, analysis pipelines and validation methods. We show that using PCR-free methods and increasing sequencing depth to ∼100 × shows benefits, as long as the tumour:control coverage ratio remains balanced. We observe widely varying mutation call rates and low concordance among analysis pipelines, reflecting the artefact-prone nature of the raw data and lack of standards for dealing with the artefacts. However, we show that, using the benchmark mutation set we have created, many issues are in fact easy to remedy and have an immediate positive impact on mutation detection accuracy.


Nature Genetics | 2016

Mutational signatures in esophageal adenocarcinoma define etiologically distinct subgroups with therapeutic relevance

Maria Secrier; Xiaodun Li; Nadeera de Silva; Matthew Eldridge; Gianmarco Contino; Jan Bornschein; Shona MacRae; Nicola Grehan; Maria O'Donovan; Ahmad Miremadi; Tsun-Po Yang; Lawrence Bower; Hamza Chettouh; Jason Crawte; Núria Galeano-Dalmau; Anna M. Grabowska; John Saunders; Timothy J. Underwood; Nicola Waddell; Andrew P. Barbour; Barbara Nutzinger; Achilleas Achilleos; Paul A.W. Edwards; Andy G. Lynch; Simon Tavaré; Rebecca C. Fitzgerald

Esophageal adenocarcinoma (EAC) has a poor outcome, and targeted therapy trials have thus far been disappointing owing to a lack of robust stratification methods. Whole-genome sequencing (WGS) analysis of 129 cases demonstrated that this is a heterogeneous cancer dominated by copy number alterations with frequent large-scale rearrangements. Co-amplification of receptor tyrosine kinases (RTKs) and/or downstream mitogenic activation is almost ubiquitous; thus tailored combination RTK inhibitor (RTKi) therapy might be required, as we demonstrate in vitro. However, mutational signatures showed three distinct molecular subtypes with potential therapeutic relevance, which we verified in an independent cohort (n = 87): (i) enrichment for BRCA signature with prevalent defects in the homologous recombination pathway; (ii) dominant T>G mutational pattern associated with a high mutational load and neoantigen burden; and (iii) C>A/T mutational pattern with evidence of an aging imprint. These subtypes could be ascertained using a clinically applicable sequencing strategy (low coverage) as a basis for therapy selection.


American Journal of Human Genetics | 2010

DNA methylome of familial breast cancer identifies distinct profiles defined by mutation status

James M. Flanagan; S Kugler; Nicola Waddell; Cn Johnstone; Anna Marsh; S Henderson; Peter T. Simpson; L. Da Silva; Kum Kum Khanna; Sunil R. Lakhani; C Boshoff; Georgia Chenevix-Trench

It is now understood that epigenetic alterations occur frequently in sporadic breast carcinogenesis, but little is known about the epigenetic alterations associated with familial breast tumors. We performed genome-wide DNA-methylation profiling on familial breast cancers (n = 33) to identify patterns of methylation specific to the different mutation groups (BRCA1, BRCA2, and BRCAx) or intrinsic subtypes of breast cancer (basal, luminal A, luminal B, HER2-amplified, and normal-like). We used methylated DNA immunoprecipitation (MeDIP) on Affymetrix promoter chips to interrogate methylation profiles across 25,500 distinct transcripts. Using a support vector machine classification algorithm, we demonstrated that genome-wide methylation profiles predicted tumor mutation status with estimated error rates of 19% (BRCA1), 31% (BRCA2), and 36% (BRCAx) but did not accurately predict the intrinsic subtypes defined by gene expression. Furthermore, using unsupervised hierarchical clustering, we identified a distinct subgroup of BRCAx tumors defined by methylation profiles. We validated these findings in the 33 tumors in the test set, as well as in an independent validation set of 47 formalin-fixed, paraffin-embedded familial breast tumors, by pyrosequencing and Epityper. Finally, gene-expression profiling and SNP CGH array previously performed on the same samples allowed full integration of methylation, gene-expression, and copy-number data sets, revealing frequent hypermethylation of genes that also displayed loss of heterozygosity, as well as of genes that show copy-number gains, providing a potential mechanism for expression dosage compensation. Together, these data show that methylation profiles for familial breast cancers are defined by the mutation status and are distinct from the intrinsic subtypes.


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.

Collaboration


Dive into the Nicola Waddell's collaboration.

Top Co-Authors

Avatar

Katia Nones

University of Queensland

View shared research outputs
Top Co-Authors

Avatar

Ann-Marie Patch

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
Top Co-Authors

Avatar

Stephen Kazakoff

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amber L. Johns

Garvan Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Oliver Holmes

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge