scispace - formally typeset
Search or ask a question
Author

James G. Kench

Bio: James G. Kench is an academic researcher from Royal Prince Alfred Hospital. The author has contributed to research in topics: Prostate cancer & Cancer. The author has an hindex of 64, co-authored 213 publications receiving 24889 citations. Previous affiliations of James G. Kench include Royal North Shore Hospital & Westmead Hospital.


Papers
More filters
Journal ArticleDOI
Peter Bailey1, David K. Chang2, Katia Nones1, Katia Nones3, Amber L. Johns4, Ann-Marie Patch1, Ann-Marie Patch3, Marie-Claude Gingras5, David Miller1, David Miller4, Angelika N. Christ1, Timothy J. C. Bruxner1, Michael C.J. Quinn1, Michael C.J. Quinn3, Craig Nourse2, Craig Nourse1, Murtaugh Lc6, Ivon Harliwong1, Senel Idrisoglu1, Suzanne Manning1, Ehsan Nourbakhsh1, Shivangi Wani3, Shivangi Wani1, J. Lynn Fink1, Oliver Holmes3, Oliver Holmes1, Chin4, Matthew J. Anderson1, Stephen H. Kazakoff1, Stephen H. Kazakoff3, Conrad Leonard1, Conrad Leonard3, Felicity Newell1, Nicola Waddell1, Scott Wood3, Scott Wood1, Qinying Xu1, Qinying Xu3, Peter J. Wilson1, Nicole Cloonan3, Nicole Cloonan1, Karin S. Kassahn7, Karin S. Kassahn8, Karin S. Kassahn1, Darrin Taylor1, Kelly Quek1, Alan J. Robertson1, Lorena Pantano9, Laura Mincarelli2, Luis Navarro Sanchez2, Lisa Evers2, Jianmin Wu4, Mark Pinese4, Mark J. Cowley4, Jones4, Jones2, Emily K. Colvin4, Adnan Nagrial4, Emily S. Humphrey4, Lorraine A. Chantrill10, Lorraine A. Chantrill4, Amanda Mawson4, Jeremy L. Humphris4, Angela Chou11, Angela Chou4, Marina Pajic4, Marina Pajic12, Christopher J. Scarlett13, Christopher J. Scarlett4, Andreia V. Pinho4, Marc Giry-Laterriere4, Ilse Rooman4, Jaswinder S. Samra14, James G. Kench15, James G. Kench4, James G. Kench16, Jessica A. Lovell4, Neil D. Merrett12, Christopher W. Toon4, Krishna Epari17, Nam Q. Nguyen18, Andrew Barbour19, Nikolajs Zeps20, Kim Moran-Jones2, Nigel B. Jamieson2, Janet Graham2, Janet Graham21, Fraser Duthie22, Karin A. Oien22, Karin A. Oien4, Hair J22, Robert Grützmann23, Anirban Maitra24, Christine A. Iacobuzio-Donahue25, Christopher L. Wolfgang26, Richard A. Morgan26, Rita T. Lawlor, Corbo, Claudio Bassi, Borislav Rusev, Paola Capelli27, Roberto Salvia, Giampaolo Tortora, Debabrata Mukhopadhyay28, Gloria M. Petersen28, Munzy Dm5, William E. Fisher5, Saadia A. Karim, Eshleman26, Ralph H. Hruban26, Christian Pilarsky23, Jennifer P. Morton, Owen J. Sansom2, Aldo Scarpa27, Elizabeth A. Musgrove2, Ulla-Maja Bailey2, Oliver Hofmann9, Oliver Hofmann2, R. L. Sutherland4, David A. Wheeler5, Anthony J. Gill4, Anthony J. Gill16, Richard A. Gibbs5, John V. Pearson1, John V. Pearson3, Andrew V. Biankin, Sean M. Grimmond2, Sean M. Grimmond1, Sean M. Grimmond29 
03 Mar 2016-Nature
TL;DR: Detailed genomic analysis of 456 pancreatic ductal adenocarcinomas identified 32 recurrently mutated genes that aggregate into 10 pathways: KRAS, TGF-β, WNT, NOTCH, ROBO/SLIT signalling, G1/S transition, SWI-SNF, chromatin modification, DNA repair and RNA processing.
Abstract: Integrated genomic analysis of 456 pancreatic ductal adenocarcinomas identified 32 recurrently mutated genes that aggregate into 10 pathways: KRAS, TGF-β, WNT, NOTCH, ROBO/SLIT signalling, G1/S transition, SWI-SNF, chromatin modification, DNA repair and RNA processing. Expression analysis defined 4 subtypes: (1) squamous; (2) pancreatic progenitor; (3) immunogenic; and (4) aberrantly differentiated endocrine exocrine (ADEX) that correlate with histopathological characteristics. Squamous tumours are enriched for TP53 and KDM6A mutations, upregulation of the TP63∆N transcriptional network, hypermethylation of pancreatic endodermal cell-fate determining genes and have a poor prognosis. Pancreatic progenitor tumours preferentially express genes involved in early pancreatic development (FOXA2/3, PDX1 and MNX1). ADEX tumours displayed upregulation of genes that regulate networks involved in KRAS activation, exocrine (NR5A2 and RBPJL), and endocrine differentiation (NEUROD1 and NKX2-2). Immunogenic tumours contained upregulated immune networks including pathways involved in acquired immune suppression. These data infer differences in the molecular evolution of pancreatic cancer subtypes and identify opportunities for therapeutic development.

2,443 citations

Journal ArticleDOI
TL;DR: A simple scoring system accurately separates patients with nonalcoholic fatty liver disease with and without advanced fibrosis, rendering liver biopsy for identification ofAdvanced fibrosis unnecessary in a substantial proportion of patients.

2,387 citations

Journal ArticleDOI
Thomas J. Hudson1, Thomas J. Hudson2, Warwick Anderson3, Axel Aretz4  +270 moreInstitutions (92)
15 Apr 2010
TL;DR: Systematic studies of more than 25,000 cancer genomes will reveal the repertoire of oncogenic mutations, uncover traces of the mutagenic influences, define clinically relevant subtypes for prognosis and therapeutic management, and enable the development of new cancer therapies.
Abstract: The International Cancer Genome Consortium (ICGC) was launched to coordinate large-scale cancer genome studies in tumours from 50 different cancer types and/or subtypes that are of clinical and societal importance across the globe. Systematic studies of more than 25,000 cancer genomes at the genomic, epigenomic and transcriptomic levels will reveal the repertoire of oncogenic mutations, uncover traces of the mutagenic influences, define clinically relevant subtypes for prognosis and therapeutic management, and enable the development of new cancer therapies.

2,041 citations

Journal ArticleDOI
Nicola Waddell1, Marina Pajic2, Ann-Marie Patch3, David K. Chang2, Karin S. Kassahn3, Peter Bailey3, Amber L. Johns2, David Miller3, Katia Nones3, Kelly Quek3, Michael C.J. Quinn3, Alan J. Robertson3, Muhammad Zaki Hidayatullah Fadlullah3, Timothy J. C. Bruxner3, Angelika N. Christ3, Ivon Harliwong3, Senel Idrisoglu3, Suzanne Manning3, Craig Nourse3, Ehsan Nourbakhsh3, Shivangi Wani3, Peter J. Wilson3, Emma Markham3, Nicole Cloonan1, Matthew J. Anderson3, J. Lynn Fink3, Oliver Holmes3, Stephen H. Kazakoff3, Conrad Leonard3, Felicity Newell3, Barsha Poudel3, Sarah Song3, Darrin Taylor3, Nick Waddell3, Scott Wood3, Qinying Xu3, Jianmin Wu2, Mark Pinese2, Mark J. Cowley2, Hong C. Lee2, Marc D. Jones2, Adnan Nagrial2, Jeremy L. Humphris2, Lorraine A. Chantrill2, Venessa T. Chin2, Angela Steinmann2, Amanda Mawson2, Emily S. Humphrey2, Emily K. Colvin2, Angela Chou2, Christopher J. Scarlett2, Andreia V. Pinho2, Marc Giry-Laterriere2, Ilse Rooman2, Jaswinder S. Samra4, James G. Kench2, Jessica A. Pettitt2, Neil D. Merrett5, Christopher W. Toon2, Krishna Epari6, Nam Q. Nguyen7, Andrew Barbour8, Nikolajs Zeps9, Nigel B. Jamieson10, Janet Graham11, Simone P. Niclou, Rolf Bjerkvig12, Robert Grützmann13, Daniela Aust13, Ralph H. Hruban14, Anirban Maitra15, Christine A. Iacobuzio-Donahue16, Christopher L. Wolfgang14, Richard A. Morgan14, Rita T. Lawlor17, Vincenzo Corbo, Claudio Bassi, Massimo Falconi, Giuseppe Zamboni17, Giampaolo Tortora, Margaret A. Tempero18, Anthony J. Gill2, James R. Eshleman14, Christian Pilarsky13, Aldo Scarpa17, Elizabeth A. Musgrove19, John V. Pearson1, Andrew V. Biankin2, Sean M. Grimmond3 
26 Feb 2015-Nature
TL;DR: Genomic instability co-segregated with inactivation of DNA maintenance genes (BRCA1, BRCA2 or PALB2) and a mutational signature of DNA damage repair deficiency, and 4 of 5 individuals with these measures of defective DNA maintenance responded to platinum therapy.
Abstract: 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.

2,035 citations

Journal ArticleDOI
Andrew V. Biankin1, Andrew V. Biankin2, Andrew V. Biankin3, Nicola Waddell4, Karin S. Kassahn4, Marie-Claude Gingras5, Lakshmi Muthuswamy6, Amber L. Johns2, David Miller4, Peter Wilson4, Ann-Marie Patch4, Jianmin Wu2, David K. Chang3, David K. Chang1, David K. Chang2, Mark J. Cowley2, Brooke Gardiner4, Sarah Song4, Ivon Harliwong4, Senel Idrisoglu4, Craig Nourse4, Ehsan Nourbakhsh4, Suzanne Manning4, Shivangi Wani4, Milena Gongora4, Marina Pajic2, Christopher J. Scarlett2, Christopher J. Scarlett7, Anthony J. Gill8, Anthony J. Gill9, Anthony J. Gill2, Andreia V. Pinho2, Ilse Rooman2, Matthew J. Anderson4, Oliver Holmes4, Conrad Leonard4, Darrin Taylor4, Scott Wood4, Qinying Xu4, Katia Nones4, J. Lynn Fink4, Angelika N. Christ4, Timothy J. C. Bruxner4, Nicole Cloonan4, Gabriel Kolle10, Felicity Newell4, Mark Pinese2, R. Scott Mead11, R. Scott Mead2, Jeremy L. Humphris2, Warren Kaplan2, Marc D. Jones2, Emily K. Colvin2, Adnan Nagrial2, Emily S. Humphrey2, Angela Chou11, Angela Chou2, Venessa T. Chin2, Lorraine A. Chantrill2, Amanda Mawson2, Jaswinder S. Samra9, James G. Kench2, James G. Kench12, James G. Kench8, Jessica A. Lovell2, Roger J. Daly2, Neil D. Merrett8, Neil D. Merrett1, Christopher W. Toon2, Krishna Epari13, Nam Q. Nguyen14, Andrew Barbour4, Nikolajs Zeps15, Nipun Kakkar5, Fengmei Zhao5, Yuan Qing Wu5, Min Wang5, Donna M. Muzny5, William E. Fisher5, F. Charles Brunicardi16, Sally E. Hodges5, Jeffrey G. Reid5, Jennifer Drummond5, Kyle Chang5, Yi Han5, Lora Lewis5, Huyen Dinh5, Christian J. Buhay5, Timothy Beck6, Lee Timms6, Michelle Sam6, Kimberly Begley6, Andrew M.K. Brown6, Deepa Pai6, Ami Panchal6, Nicholas Buchner6, Richard de Borja6, Robert E. Denroche6, Christina K. Yung6, Stefano Serra17, Nicole Onetto6, Debabrata Mukhopadhyay18, Ming-Sound Tsao17, Patricia Shaw17, Gloria M. Petersen18, Steven Gallinger17, Steven Gallinger19, Ralph H. Hruban20, Anirban Maitra20, Christine A. Iacobuzio-Donahue20, Richard D. Schulick20, Christopher L. Wolfgang20, Richard A. Morgan20, Rita T. Lawlor, Paola Capelli21, Vincenzo Corbo, Maria Scardoni21, Giampaolo Tortora, Margaret A. Tempero22, Karen M. Mann23, Nancy A. Jenkins23, Pedro A. Perez-Mancera24, David J. Adams25, David A. Largaespada26, Lodewyk F. A. Wessels27, Alistair G. Rust25, Lincoln Stein6, David A. Tuveson24, Neal G. Copeland23, Elizabeth A. Musgrove2, Elizabeth A. Musgrove3, Aldo Scarpa21, James R. Eshleman20, Thomas J. Hudson6, Robert L. Sutherland2, Robert L. Sutherland3, David A. Wheeler5, John V. Pearson4, John Douglas Mcpherson6, Richard A. Gibbs5, Sean M. Grimmond4 
15 Nov 2012-Nature
TL;DR: It is found that frequent and diverse somatic aberrations in genes described traditionally as embryonic regulators of axon guidance, particularly SLIT/ROBO signalling, are also evident in murine Sleeping Beauty transposon-mediated somatic mutagenesis models of pancreatic cancer, providing further supportive evidence for the potential involvement ofAxon guidance genes in pancreatic carcinogenesis.
Abstract: Pancreatic cancer is a highly lethal malignancy with few effective therapies. We performed exome sequencing and copy number analysis to define genomic aberrations in a prospectively accrued clinical cohort (n = 142) of early (stage I and II) sporadic pancreatic ductal adenocarcinoma. Detailed analysis of 99 informative tumours identified substantial heterogeneity with 2,016 non-silent mutations and 1,628 copy-number variations. We define 16 significantly mutated genes, reaffirming known mutations (KRAS, TP53, CDKN2A, SMAD4, MLL3, TGFBR2, ARID1A and SF3B1), and uncover novel mutated genes including additional genes involved in chromatin modification (EPC1 and ARID2), DNA damage repair (ATM) and other mechanisms (ZIM2, MAP2K4, NALCN, SLC16A4 and MAGEA6). Integrative analysis with in vitro functional data and animal models provided supportive evidence for potential roles for these genetic aberrations in carcinogenesis. Pathway-based analysis of recurrently mutated genes recapitulated clustering in core signalling pathways in pancreatic ductal adenocarcinoma, and identified new mutated genes in each pathway. We also identified frequent and diverse somatic aberrations in genes described traditionally as embryonic regulators of axon guidance, particularly SLIT/ROBO signalling, which was also evident in murine Sleeping Beauty transposon-mediated somatic mutagenesis models of pancreatic cancer, providing further supportive evidence for the potential involvement of axon guidance genes in pancreatic carcinogenesis.

1,752 citations


Cited by
More filters
Journal ArticleDOI
TL;DR: The cBio Cancer Genomics Portal significantly lowers the barriers between complex genomic data and cancer researchers who want rapid, intuitive, and high-quality access to molecular profiles and clinical attributes from large-scale cancer genomics projects and empowers researchers to translate these rich data sets into biologic insights and clinical applications.
Abstract: The cBio Cancer Genomics Portal (http://cbioportal.org) is an open-access resource for interactive exploration of multidimensional cancer genomics data sets, currently providing access to data from more than 5,000 tumor samples from 20 cancer studies. The cBio Cancer Genomics Portal significantly lowers the barriers between complex genomic data and cancer researchers who want rapid, intuitive, and high-quality access to molecular profiles and clinical attributes from large-scale cancer genomics projects and empowers researchers to translate these rich data sets into biologic insights and clinical applications.

11,912 citations

Journal ArticleDOI
TL;DR: A practical guide to the analysis and visualization features of the cBioPortal for Cancer Genomics, which makes complex cancer genomics profiles accessible to researchers and clinicians without requiring bioinformatics expertise, thus facilitating biological discoveries.
Abstract: The cBioPortal for Cancer Genomics (http://cbioportal.org) provides a Web resource for exploring, visualizing, and analyzing multidimensional cancer genomics data. The portal reduces molecular profiling data from cancer tissues and cell lines into readily understandable genetic, epigenetic, gene expression, and proteomic events. The query interface combined with customized data storage enables researchers to interactively explore genetic alterations across samples, genes, and pathways and, when available in the underlying data, to link these to clinical outcomes. The portal provides graphical summaries of gene-level data from multiple platforms, network visualization and analysis, survival analysis, patient-centric queries, and software programmatic access. The intuitive Web interface of the portal makes complex cancer genomics profiles accessible to researchers and clinicians without requiring bioinformatics expertise, thus facilitating biological discoveries. Here, we provide a practical guide to the analysis and visualization features of the cBioPortal for Cancer Genomics.

10,947 citations

Journal ArticleDOI
Ludmil B. Alexandrov1, Serena Nik-Zainal2, Serena Nik-Zainal3, David C. Wedge1, Samuel Aparicio4, Sam Behjati1, Sam Behjati5, Andrew V. Biankin, Graham R. Bignell1, Niccolo Bolli1, Niccolo Bolli5, Åke Borg2, Anne Lise Børresen-Dale6, Anne Lise Børresen-Dale7, Sandrine Boyault8, Birgit Burkhardt8, Adam Butler1, Carlos Caldas9, Helen Davies1, Christine Desmedt, Roland Eils5, Jorunn E. Eyfjord10, John A. Foekens11, Mel Greaves12, Fumie Hosoda13, Barbara Hutter5, Tomislav Ilicic1, Sandrine Imbeaud14, Sandrine Imbeaud15, Marcin Imielinsk14, Natalie Jäger5, David T. W. Jones16, David T. Jones1, Stian Knappskog17, Stian Knappskog11, Marcel Kool11, Sunil R. Lakhani18, Carlos López-Otín18, Sancha Martin1, Nikhil C. Munshi19, Nikhil C. Munshi20, Hiromi Nakamura13, Paul A. Northcott16, Marina Pajic21, Elli Papaemmanuil1, Angelo Paradiso22, John V. Pearson23, Xose S. Puente18, Keiran Raine1, Manasa Ramakrishna1, Andrea L. Richardson22, Andrea L. Richardson20, Julia Richter22, Philip Rosenstiel22, Matthias Schlesner5, Ton N. Schumacher24, Paul N. Span25, Jon W. Teague1, Yasushi Totoki13, Andrew Tutt24, Rafael Valdés-Mas18, Marit M. van Buuren25, Laura van ’t Veer26, Anne Vincent-Salomon27, Nicola Waddell23, Lucy R. Yates1, Icgc PedBrain24, Jessica Zucman-Rossi14, Jessica Zucman-Rossi15, P. Andrew Futreal1, Ultan McDermott1, Peter Lichter24, Matthew Meyerson14, Matthew Meyerson20, Sean M. Grimmond23, Reiner Siebert22, Elias Campo28, Tatsuhiro Shibata13, Stefan M. Pfister11, Stefan M. Pfister16, Peter J. Campbell29, Peter J. Campbell3, Peter J. Campbell30, Michael R. Stratton31, Michael R. Stratton3 
22 Aug 2013-Nature
TL;DR: It is shown that hypermutation localized to small genomic regions, ‘kataegis’, is found in many cancer types, and this results reveal the diversity of mutational processes underlying the development of cancer.
Abstract: 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.

7,904 citations

01 Jan 2010
TL;DR: Since the publication of the American Association for the Study of Liver Diseases (AASLD) practice guidelines on the management of hepatocellular carcinoma (HCC) in 2005, new information has emerged that requires that the guidelines be updated.
Abstract: Since the publication of the American Association for the Study of Liver Diseases (AASLD) practice guidelines on the management of hepatocellular carcinoma (HCC) in 2005, new information has emerged that requires that the guidelines be updated. The full version of the new guidelines is available on the AASLD Web site at http://www.aasld.org/practiceguidelines/ Documents/Bookmarked%20Practice%20Guidelines/ HCCUpdate2010.pdf. Here, we briefly describe only new or changed recommendations.

6,642 citations

Journal ArticleDOI
29 Mar 2013-Science
TL;DR: This work has revealed the genomic landscapes of common forms of human cancer, which consists of a small number of “mountains” (genes altered in a high percentage of tumors) and a much larger number of "hills" (Genes altered infrequently).
Abstract: Over the past decade, comprehensive sequencing efforts have revealed the genomic landscapes of common forms of human cancer. For most cancer types, this landscape consists of a small number of “mountains” (genes altered in a high percentage of tumors) and a much larger number of “hills” (genes altered infrequently). To date, these studies have revealed ~140 genes that, when altered by intragenic mutations, can promote or “drive” tumorigenesis. A typical tumor contains two to eight of these “driver gene” mutations; the remaining mutations are passengers that confer no selective growth advantage. Driver genes can be classified into 12 signaling pathways that regulate three core cellular processes: cell fate, cell survival, and genome maintenance. A better understanding of these pathways is one of the most pressing needs in basic cancer research. Even now, however, our knowledge of cancer genomes is sufficient to guide the development of more effective approaches for reducing cancer morbidity and mortality.

6,441 citations