scispace - formally typeset
Search or ask a question
Author

Michael C.J. Quinn

Bio: Michael C.J. Quinn is an academic researcher from QIMR Berghofer Medical Research Institute. The author has contributed to research in topics: Serous fluid & Transcriptome. The author has an hindex of 19, co-authored 37 publications receiving 6747 citations. Previous affiliations of Michael C.J. Quinn include Université de Montréal & McGill University.

Papers
More filters
Journal ArticleDOI
Peter Bailey1, David K. Chang2, Katia Nones1, Katia Nones3, Amber L. Johns4, Ann-Marie Patch3, Ann-Marie Patch1, Marie-Claude Gingras5, David Miller4, David Miller1, Angelika N. Christ1, Timothy J. C. Bruxner1, Michael C.J. Quinn1, Michael C.J. Quinn3, Craig Nourse1, Craig Nourse2, 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 Leonard3, Conrad Leonard1, Felicity Newell1, Nicola Waddell1, Scott Wood3, Scott Wood1, Qinying Xu1, Qinying Xu3, Peter J. Wilson1, Nicole Cloonan3, Nicole Cloonan1, Karin S. Kassahn7, Karin S. Kassahn1, Karin S. Kassahn8, 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 Chou4, Angela Chou11, 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 Graham21, Janet Graham2, 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. Gill16, Richard A. Gibbs5, John V. Pearson1, John V. Pearson3, Andrew V. Biankin, Sean M. Grimmond1, Sean M. Grimmond29, Sean M. Grimmond2 
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
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 Bailey1, Karin S. Kassahn1, Karin S. Kassahn6, Felicity Newell1, Michael C.J. Quinn2, Michael C.J. Quinn1, Stephen H. Kazakoff2, Stephen H. Kazakoff1, Kelly Quek1, 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. Quinn1, 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 Miller1, Gisela Mir Arnau3, Richard W. Tothill3, Timothy P. Holloway3, Timothy Semple3, Ivon Harliwong1, Craig Nourse1, Ehsan Nourbakhsh1, Suzanne Manning1, Senel Idrisoglu1, Timothy J. C. Bruxner1, Angelika N. Christ1, Barsha Poudel1, Oliver Holmes2, Oliver Holmes1, Matthew J. Anderson1, Conrad Leonard1, Conrad Leonard2, Andrew Lonie11, Nathan E. Hall12, Scott Wood2, Scott Wood1, Darrin Taylor1, Qinying Xu1, Qinying Xu2, J. Lynn Fink1, Nick Waddell1, Ronny Drapkin13, Euan A. Stronach7, Hani Gabra7, Robert S. Brown7, A. Jewell14, Shivashankar H. Nagaraj1, Emma Markham1, Peter Wilson1, Jason Ellul3, Orla McNally9, Maria A. Doyle3, Ravikiran Vedururu3, Collin Stewart15, Ernst Lengyel14, John V. Pearson1, John V. Pearson2, Nicola Waddell2, Nicola Waddell1, Anna deFazio9, Sean M. Grimmond5, Sean M. Grimmond1, David D.L. Bowtell4, David D.L. Bowtell7, 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

Journal ArticleDOI
Aldo Scarpa, David K. Chang, Katia Nones1, Katia Nones2, Vincenzo Corbo, Ann-Marie Patch2, Ann-Marie Patch1, Peter Bailey3, Peter Bailey2, Rita T. Lawlor, Amber L. Johns4, David Miller2, Andrea Mafficini, Borislav Rusev, Maria Scardoni, Davide Antonello, Stefano Barbi, Katarzyna O. Sikora, Sara Cingarlini, Caterina Vicentini, Skye McKay4, Michael C.J. Quinn1, Michael C.J. Quinn2, Timothy J. C. Bruxner2, Angelika N. Christ2, Ivon Harliwong2, Senel Idrisoglu2, Suzanne McLean2, Craig Nourse3, Craig Nourse2, Ehsan Nourbakhsh2, Peter J. Wilson2, Matthew J. Anderson2, J. Lynn Fink2, Felicity Newell2, Felicity Newell1, Nick Waddell2, Oliver Holmes1, Oliver Holmes2, Stephen H. Kazakoff1, Stephen H. Kazakoff2, Conrad Leonard1, Conrad Leonard2, Scott Wood1, Scott Wood2, Qinying Xu2, Qinying Xu1, Shivashankar H. Nagaraj2, Eliana Amato, Irene Dalai, Samantha Bersani, Ivana Cataldo, Angelo Paolo Dei Tos5, Paola Capelli, Maria Vittoria Davì, Luca Landoni, Anna Malpaga, Marco Miotto, Vicki L. J. Whitehall1, Vicki L. J. Whitehall2, Barbara A. Leggett6, Barbara A. Leggett1, Barbara A. Leggett2, Janelle L. Harris1, Jonathan M. Harris7, Marc D. Jones3, Jeremy L. Humphris4, Lorraine A. Chantrill4, Venessa T. Chin4, Adnan Nagrial4, Marina Pajic4, Christopher J. Scarlett8, Christopher J. Scarlett4, Andreia V. Pinho4, Ilse Rooman4, Christopher W. Toon4, Jianmin Wu4, Jianmin Wu9, Mark Pinese4, Mark J. Cowley4, Andrew Barbour10, Amanda Mawson4, Emily S. Humphrey4, Emily K. Colvin4, Angela Chou4, Angela Chou11, Jessica A. Lovell4, Nigel B. Jamieson3, Nigel B. Jamieson12, Fraser Duthie3, Marie-Claude Gingras13, Marie-Claude Gingras14, William E. Fisher14, Rebecca A. Dagg15, Loretta Lau15, Michael Lee16, Hilda A. Pickett16, Roger R. Reddel16, Jaswinder S. Samra17, Jaswinder S. Samra18, James G. Kench4, James G. Kench19, James G. Kench17, Neil D. Merrett17, Neil D. Merrett20, Krishna Epari21, Nam Q. Nguyen22, Nikolajs Zeps23, Nikolajs Zeps24, Massimo Falconi, Michele Simbolo, Giovanni Butturini, George Van Buren14, Stefano Partelli, Matteo Fassan, Kum Kum Khanna1, Anthony J. Gill17, Anthony J. Gill4, David A. Wheeler13, Richard A. Gibbs13, Elizabeth A. Musgrove3, Claudio Bassi, Giampaolo Tortora, Paolo Pederzoli, John V. Pearson1, John V. Pearson2, Nicola Waddell2, Nicola Waddell1, Andrew V. Biankin, Sean M. Grimmond25 
02 Mar 2017-Nature
TL;DR: In this paper, the authors performed whole-genome sequencing of 102 primary pancreatic neuroendocrine tumours and defined the genomic events that characterize their pathogenesis, including a deficiency in G:C,>T:A base excision repair due to inactivation of MUTYH, which encodes a DNA glycosylase.
Abstract: The diagnosis of pancreatic neuroendocrine tumours (PanNETs) is increasing owing to more sensitive detection methods, and this increase is creating challenges for clinical management. We performed whole-genome sequencing of 102 primary PanNETs and defined the genomic events that characterize their pathogenesis. Here we describe the mutational signatures they harbour, including a deficiency in G:C > T:A base excision repair due to inactivation of MUTYH, which encodes a DNA glycosylase. Clinically sporadic PanNETs contain a larger-than-expected proportion of germline mutations, including previously unreported mutations in the DNA repair genes MUTYH, CHEK2 and BRCA2. Together with mutations in MEN1 and VHL, these mutations occur in 17% of patients. Somatic mutations, including point mutations and gene fusions, were commonly found in genes involved in four main pathways: chromatin remodelling, DNA damage repair, activation of mTOR signalling (including previously undescribed EWSR1 gene fusions), and telomere maintenance. In addition, our gene expression analyses identified a subgroup of tumours associated with hypoxia and HIF signalling.

637 citations


Cited by
More filters
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 Nones3, Katia Nones1, Amber L. Johns4, Ann-Marie Patch3, Ann-Marie Patch1, Marie-Claude Gingras5, David Miller4, David Miller1, 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 Wani1, Shivangi Wani3, 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 Wood3, Scott Wood1, Qinying Xu3, Qinying Xu1, Peter J. Wilson1, Nicole Cloonan1, Nicole Cloonan3, Karin S. Kassahn1, Karin S. Kassahn7, Karin S. Kassahn8, 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 Chou4, Angela Chou11, 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. Kench16, James G. Kench4, 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 Graham21, Janet Graham2, 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. 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
TL;DR: Inflammation is a biological response of the immune system that can be triggered by a variety of factors, including pathogens, damaged cells and toxic compounds, potentially leading to tissue damage or disease.
Abstract: Inflammation is a biological response of the immune system that can be triggered by a variety of factors, including pathogens, damaged cells and toxic compounds. These factors may induce acute and/or chronic inflammatory responses in the heart, pancreas, liver, kidney, lung, brain, intestinal tract and reproductive system, potentially leading to tissue damage or disease. Both infectious and non-infectious agents and cell damage activate inflammatory cells and trigger inflammatory signaling pathways, most commonly the NF-κB, MAPK, and JAK-STAT pathways. Here, we review inflammatory responses within organs, focusing on the etiology of inflammation, inflammatory response mechanisms, resolution of inflammation, and organ-specific inflammatory responses.

2,197 citations

01 Jan 2011
TL;DR: The sheer volume and scope of data posed by this flood of data pose a significant challenge to the development of efficient and intuitive visualization tools able to scale to very large data sets and to flexibly integrate multiple data types, including clinical data.
Abstract: Rapid improvements in sequencing and array-based platforms are resulting in a flood of diverse genome-wide data, including data from exome and whole-genome sequencing, epigenetic surveys, expression profiling of coding and noncoding RNAs, single nucleotide polymorphism (SNP) and copy number profiling, and functional assays. Analysis of these large, diverse data sets holds the promise of a more comprehensive understanding of the genome and its relation to human disease. Experienced and knowledgeable human review is an essential component of this process, complementing computational approaches. This calls for efficient and intuitive visualization tools able to scale to very large data sets and to flexibly integrate multiple data types, including clinical data. However, the sheer volume and scope of data pose a significant challenge to the development of such tools.

2,187 citations

25 May 2011
TL;DR: A quantitative analysis of the timing of the genetic evolution of pancreatic cancer was performed, indicating at least a decade between the occurrence of the initiating mutation and the birth of the parental, non-metastatic founder cell.
Abstract: Metastasis, the dissemination and growth of neoplastic cells in an organ distinct from that in which they originated, is the most common cause of death in cancer patients. This is particularly true for pancreatic cancers, where most patients are diagnosed with metastatic disease and few show a sustained response to chemotherapy or radiation therapy. Whether the dismal prognosis of patients with pancreatic cancer compared to patients with other types of cancer is a result of late diagnosis or early dissemination of disease to distant organs is not known. Here we rely on data generated by sequencing the genomes of seven pancreatic cancer metastases to evaluate the clonal relationships among primary and metastatic cancers. We find that clonal populations that give rise to distant metastases are represented within the primary carcinoma, but these clones are genetically evolved from the original parental, non-metastatic clone. Thus, genetic heterogeneity of metastases reflects that within the primary carcinoma. A quantitative analysis of the timing of the genetic evolution of pancreatic cancer was performed, indicating at least a decade between the occurrence of the initiating mutation and the birth of the parental, non-metastatic founder cell. At least five more years are required for the acquisition of metastatic ability and patients die an average of two years thereafter. These data provide novel insights into the genetic features underlying pancreatic cancer progression and define a broad time window of opportunity for early detection to prevent deaths from metastatic disease.

2,019 citations