scispace - formally typeset
Search or ask a question
Author

Oliver Holmes

Bio: Oliver Holmes is an academic researcher from QIMR Berghofer Medical Research Institute. The author has contributed to research in topics: Medicine & Germline mutation. The author has an hindex of 24, co-authored 41 publications receiving 9645 citations. Previous affiliations of Oliver Holmes include Harvard University & Foundation Medicine.


Papers
More filters
Journal ArticleDOI
Peter Bailey1, David K. Chang2, Katia Nones3, Katia Nones1, Amber L. Johns4, Ann-Marie Patch3, Ann-Marie Patch1, 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. Kazakoff3, Stephen H. Kazakoff1, Conrad Leonard1, Conrad Leonard3, Felicity Newell1, Nicola Waddell1, Scott Wood3, Scott Wood1, Qinying Xu3, Qinying Xu1, Peter J. Wilson1, Nicole Cloonan1, Nicole Cloonan3, 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, Jones2, Jones4, Emily K. Colvin4, Adnan Nagrial4, Emily S. Humphrey4, Lorraine A. Chantrill10, Lorraine A. Chantrill4, Amanda Mawson4, Jeremy L. Humphris4, Angela Chou11, Angela Chou4, Marina Pajic12, Marina Pajic4, 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. Oien4, Karin A. Oien22, 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 Hofmann2, Oliver Hofmann9, R. L. Sutherland4, David A. Wheeler5, Anthony J. Gill4, Anthony J. Gill15, Richard A. Gibbs5, John V. Pearson3, John V. Pearson1, Andrew V. Biankin, Sean M. Grimmond1, Sean M. Grimmond2, 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
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. Chang1, David K. Chang2, David K. Chang3, 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. Scarlett7, Christopher J. Scarlett2, Anthony J. Gill8, Anthony J. Gill2, Anthony J. Gill9, 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 Chou2, Angela Chou11, Venessa T. Chin2, Lorraine A. Chantrill2, Amanda Mawson2, Jaswinder S. Samra8, James G. Kench12, James G. Kench2, James G. Kench9, Jessica A. Lovell2, Roger J. Daly2, Neil D. Merrett1, Neil D. Merrett9, 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. Musgrove3, Elizabeth A. Musgrove2, 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

Journal ArticleDOI
Peter J. Campbell1, Gad Getz2, Jan O. Korbel3, Joshua M. Stuart4  +1329 moreInstitutions (238)
06 Feb 2020-Nature
TL;DR: The flagship paper of the ICGC/TCGA Pan-Cancer Analysis of Whole Genomes Consortium describes the generation of the integrative analyses of 2,658 whole-cancer genomes and their matching normal tissues across 38 tumour types, the structures for international data sharing and standardized analyses, and the main scientific findings from across the consortium studies.
Abstract: Cancer is driven by genetic change, and the advent of massively parallel sequencing has enabled systematic documentation of this variation at the whole-genome scale1,2,3. Here we report the integrative analysis of 2,658 whole-cancer genomes and their matching normal tissues across 38 tumour types from the Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium of the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA). We describe the generation of the PCAWG resource, facilitated by international data sharing using compute clouds. On average, cancer genomes contained 4–5 driver mutations when combining coding and non-coding genomic elements; however, in around 5% of cases no drivers were identified, suggesting that cancer driver discovery is not yet complete. Chromothripsis, in which many clustered structural variants arise in a single catastrophic event, is frequently an early event in tumour evolution; in acral melanoma, for example, these events precede most somatic point mutations and affect several cancer-associated genes simultaneously. Cancers with abnormal telomere maintenance often originate from tissues with low replicative activity and show several mechanisms of preventing telomere attrition to critical levels. Common and rare germline variants affect patterns of somatic mutation, including point mutations, structural variants and somatic retrotransposition. A collection of papers from the PCAWG Consortium describes non-coding mutations that drive cancer beyond those in the TERT promoter4; identifies new signatures of mutational processes that cause base substitutions, small insertions and deletions and structural variation5,6; analyses timings and patterns of tumour evolution7; describes the diverse transcriptional consequences of somatic mutation on splicing, expression levels, fusion genes and promoter activity8,9; and evaluates a range of more-specialized features of cancer genomes8,10,11,12,13,14,15,16,17,18.

1,600 citations

Journal ArticleDOI
Ann-Marie Patch1, Ann-Marie Patch2, Elizabeth L. Christie3, Dariush Etemadmoghadam4, Dariush Etemadmoghadam3, Dale W. Garsed3, Joshy George, Sian Fereday3, Katia Nones1, Katia Nones2, Prue A. Cowin3, Kathryn Alsop3, Peter Bailey5, Peter Bailey2, Karin S. Kassahn2, Karin S. Kassahn6, Felicity Newell2, Michael C.J. Quinn2, Michael C.J. Quinn1, Stephen H. Kazakoff1, Stephen H. Kazakoff2, Kelly Quek2, Charlotte Wilhelm-Benartzi7, Edward Curry7, Huei San Leong3, Anne Hamilton4, Anne Hamilton8, Anne Hamilton3, Linda Mileshkin3, George Au-Yeung3, Catherine Kennedy9, Jillian Hung9, Yoke Eng Chiew9, Paul R. Harnett9, Michael Friedlander10, Michael C. J. Quinn2, Jan Pyman8, Stephen Cordner, Patricia C. M. O’Brien, Jodie Leditschke, Greg Young, Kate Strachan, Paul Waring4, Walid J Azar3, Chris Mitchell3, Nadia Traficante3, Joy Hendley3, Heather Thorne3, Mark Shackleton3, David Miller2, Gisela Mir Arnau3, Richard W. Tothill3, Timothy P. Holloway3, Timothy Semple3, Ivon Harliwong2, Craig Nourse2, Ehsan Nourbakhsh2, Suzanne Manning2, Senel Idrisoglu2, Timothy J. C. Bruxner2, Angelika N. Christ2, Barsha Poudel2, Oliver Holmes2, Oliver Holmes1, Matthew J. Anderson2, Conrad Leonard1, Conrad Leonard2, Andrew Lonie11, Nathan E. Hall12, Scott Wood2, Scott Wood1, Darrin Taylor2, Qinying Xu2, Qinying Xu1, J. Lynn Fink2, Nick Waddell2, Ronny Drapkin13, Euan A. Stronach7, Hani Gabra7, Robert S. Brown7, A. Jewell14, Shivashankar H. Nagaraj2, Emma Markham2, Peter Wilson2, Jason Ellul3, Orla McNally9, Maria A. Doyle3, Ravikiran Vedururu3, Collin Stewart15, Ernst Lengyel14, John V. Pearson1, John V. Pearson2, Nicola Waddell1, Nicola Waddell2, Anna deFazio9, Sean M. Grimmond2, Sean M. Grimmond5, David D.L. Bowtell7, David D.L. Bowtell4, David D.L. Bowtell3 
28 May 2015-Nature
TL;DR: It is shown that gene breakage commonly inactivates the tumour suppressors RB1, NF1, RAD51B and PTEN in HGSC, and contributes to acquired chemotherapy resistance.
Abstract: 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.

1,195 citations


Cited by
More filters
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

Journal ArticleDOI
TL;DR: The Reactome Knowledgebase provides molecular details of signal transduction, transport, DNA replication, metabolism and other cellular processes as an ordered network of molecular transformations—an extended version of a classic metabolic map, in a single consistent data model.
Abstract: The Reactome Knowledgebase (www.reactome.org) provides molecular details of signal transduction, transport, DNA replication, metabolism and other cellular processes as an ordered network of molecular transformations-an extended version of a classic metabolic map, in a single consistent data model. Reactome functions both as an archive of biological processes and as a tool for discovering unexpected functional relationships in data such as gene expression pattern surveys or somatic mutation catalogues from tumour cells. Over the last two years we redeveloped major components of the Reactome web interface to improve usability, responsiveness and data visualization. A new pathway diagram viewer provides a faster, clearer interface and smooth zooming from the entire reaction network to the details of individual reactions. Tool performance for analysis of user datasets has been substantially improved, now generating detailed results for genome-wide expression datasets within seconds. The analysis module can now be accessed through a RESTFul interface, facilitating its inclusion in third party applications. A new overview module allows the visualization of analysis results on a genome-wide Reactome pathway hierarchy using a single screen page. The search interface now provides auto-completion as well as a faceted search to narrow result lists efficiently.

5,065 citations

Journal ArticleDOI
17 Apr 2018-Immunity
TL;DR: An extensive immunogenomic analysis of more than 10,000 tumors comprising 33 diverse cancer types by utilizing data compiled by TCGA identifies six immune subtypes that encompass multiple cancer types and are hypothesized to define immune response patterns impacting prognosis.

3,246 citations

Journal ArticleDOI
Peter Bailey1, David K. Chang2, Katia Nones1, Katia Nones3, Amber L. Johns4, Ann-Marie Patch1, Ann-Marie Patch3, Marie-Claude Gingras5, David Miller4, David Miller1, Angelika N. Christ1, Timothy J. C. Bruxner1, Michael C.J. Quinn3, Michael C.J. Quinn1, Craig Nourse1, Craig Nourse2, Murtaugh Lc6, Ivon Harliwong1, Senel Idrisoglu1, Suzanne Manning1, Ehsan Nourbakhsh1, Shivangi Wani3, Shivangi Wani1, J. Lynn Fink1, Oliver Holmes1, Oliver Holmes3, Chin4, Matthew J. Anderson1, Stephen H. Kazakoff3, Stephen H. Kazakoff1, Conrad Leonard1, Conrad Leonard3, Felicity Newell1, Nicola Waddell1, Scott Wood1, Scott Wood3, Qinying Xu3, Qinying Xu1, 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, Jones2, Jones4, Emily K. Colvin4, Adnan Nagrial4, Emily S. Humphrey4, Lorraine A. Chantrill4, Lorraine A. Chantrill10, Amanda Mawson4, Jeremy L. Humphris4, Angela Chou11, Angela Chou4, Marina Pajic4, Marina Pajic12, Christopher J. Scarlett4, Christopher J. Scarlett13, Andreia V. Pinho4, Marc Giry-Laterriere4, Ilse Rooman4, Jaswinder S. Samra14, James G. Kench4, James G. Kench15, 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. Oien4, Karin A. Oien22, 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. Gill15, 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
Robert M. Samstein1, Chung-Han Lee1, Chung-Han Lee2, Alexander N. Shoushtari2, Alexander N. Shoushtari1, Matthew D. Hellmann1, Matthew D. Hellmann2, Ronglai Shen1, Yelena Y. Janjigian1, Yelena Y. Janjigian2, David Barron1, Ahmet Zehir1, Emmet Jordan1, Antonio Omuro1, Thomas Kaley1, Sviatoslav M. Kendall1, Robert J. Motzer1, Robert J. Motzer2, A. Ari Hakimi1, Martin H. Voss2, Martin H. Voss1, Paul Russo1, Jonathan E. Rosenberg2, Jonathan E. Rosenberg1, Gopa Iyer2, Gopa Iyer1, Bernard H. Bochner1, Dean F. Bajorin2, Dean F. Bajorin1, Hikmat Al-Ahmadie1, Jamie E. Chaft2, Jamie E. Chaft1, Charles M. Rudin1, Charles M. Rudin2, Gregory J. Riely2, Gregory J. Riely1, Shrujal S. Baxi2, Shrujal S. Baxi1, Alan L. Ho2, Alan L. Ho1, Richard J. Wong1, David G. Pfister2, David G. Pfister1, Jedd D. Wolchok1, Jedd D. Wolchok2, Christopher A. Barker1, Philip H. Gutin1, Cameron Brennan1, Viviane Tabar1, Ingo K. Mellinghoff1, Lisa M. DeAngelis1, Charlotte E. Ariyan1, Nancy Y. Lee1, William D. Tap1, William D. Tap2, Mrinal M. Gounder2, Mrinal M. Gounder1, Sandra P. D'Angelo1, Sandra P. D'Angelo2, Leonard B. Saltz1, Leonard B. Saltz2, Zsofia K. Stadler2, Zsofia K. Stadler1, Howard I. Scher1, Howard I. Scher2, José Baselga2, José Baselga1, Pedram Razavi2, Pedram Razavi1, Christopher A. Klebanoff2, Christopher A. Klebanoff1, Rona Yaeger2, Rona Yaeger1, Neil H. Segal2, Neil H. Segal1, Geoffrey Y. Ku2, Geoffrey Y. Ku1, Ronald P. DeMatteo1, Marc Ladanyi1, Naiyer A. Rizvi3, Michael F. Berger1, Nadeem Riaz1, David B. Solit1, Timothy A. Chan1, Luc G. T. Morris1 
TL;DR: Analysis of advanced cancer patients treated with immune-checkpoint inhibitors shows that tumor mutational burden, as assessed by targeted next-generation sequencing, predicts survival after immunotherapy across multiple cancer types.
Abstract: Immune checkpoint inhibitor (ICI) treatments benefit some patients with metastatic cancers, but predictive biomarkers are needed. Findings in selected cancer types suggest that tumor mutational burden (TMB) may predict clinical response to ICI. To examine this association more broadly, we analyzed the clinical and genomic data of 1,662 advanced cancer patients treated with ICI, and 5,371 non-ICI-treated patients, whose tumors underwent targeted next-generation sequencing (MSK-IMPACT). Among all patients, higher somatic TMB (highest 20% in each histology) was associated with better overall survival. For most cancer histologies, an association between higher TMB and improved survival was observed. The TMB cutpoints associated with improved survival varied markedly between cancer types. These data indicate that TMB is associated with improved survival in patients receiving ICI across a wide variety of cancer types, but that there may not be one universal definition of high TMB.

2,343 citations