scispace - formally typeset
Search or ask a question
Author

Nigel B. Jamieson

Bio: Nigel B. Jamieson is an academic researcher from Glasgow Royal Infirmary. The author has contributed to research in topics: Pancreatic cancer & Medicine. The author has an hindex of 44, co-authored 131 publications receiving 10913 citations. Previous affiliations of Nigel B. Jamieson include Royal North Shore Hospital & Johns Hopkins 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 Miller1, David Miller4, 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. Kazakoff1, Stephen H. Kazakoff3, Conrad Leonard3, Conrad Leonard1, 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, 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. 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. Gill16, Anthony J. Gill4, Richard A. Gibbs5, John V. Pearson3, John V. Pearson1, Andrew V. Biankin, Sean M. Grimmond29, Sean M. Grimmond1, 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
23 Nov 2017-Nature
TL;DR: In this paper, the authors used genetic, immunohistochemical and transcriptional immunoprofiling, computational biophysics, and functional assays to identify T-cell antigens in long-term survivors of pancreatic cancer.
Abstract: Pancreatic ductal adenocarcinoma is a lethal cancer with fewer than 7% of patients surviving past 5 years. T-cell immunity has been linked to the exceptional outcome of the few long-term survivors, yet the relevant antigens remain unknown. Here we use genetic, immunohistochemical and transcriptional immunoprofiling, computational biophysics, and functional assays to identify T-cell antigens in long-term survivors of pancreatic cancer. Using whole-exome sequencing and in silico neoantigen prediction, we found that tumours with both the highest neoantigen number and the most abundant CD8+ T-cell infiltrates, but neither alone, stratified patients with the longest survival. Investigating the specific neoantigen qualities promoting T-cell activation in long-term survivors, we discovered that these individuals were enriched in neoantigen qualities defined by a fitness model, and neoantigens in the tumour antigen MUC16 (also known as CA125). A neoantigen quality fitness model conferring greater immunogenicity to neoantigens with differential presentation and homology to infectious disease-derived peptides identified long-term survivors in two independent datasets, whereas a neoantigen quantity model ascribing greater immunogenicity to increasing neoantigen number alone did not. We detected intratumoural and lasting circulating T-cell reactivity to both high-quality and MUC16 neoantigens in long-term survivors of pancreatic cancer, including clones with specificity to both high-quality neoantigens and predicted cross-reactive microbial epitopes, consistent with neoantigen molecular mimicry. Notably, we observed selective loss of high-quality and MUC16 neoantigenic clones on metastatic progression, suggesting neoantigen immunoediting. Our results identify neoantigens with unique qualities as T-cell targets in pancreatic ductal adenocarcinoma. More broadly, we identify neoantigen quality as a biomarker for immunogenic tumours that may guide the application of immunotherapies.

774 citations

Journal ArticleDOI
Aldo Scarpa, David K. Chang, Katia Nones1, Katia Nones2, Vincenzo Corbo, Ann-Marie Patch1, Ann-Marie Patch2, 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. Quinn2, Michael C.J. Quinn1, Timothy J. C. Bruxner2, Angelika N. Christ2, Ivon Harliwong2, Senel Idrisoglu2, Suzanne McLean2, Craig Nourse2, Craig Nourse3, Ehsan Nourbakhsh2, Peter J. Wilson2, Matthew J. Anderson2, J. Lynn Fink2, Felicity Newell1, Felicity Newell2, Nick Waddell2, Oliver Holmes2, Oliver Holmes1, Stephen H. Kazakoff2, Stephen H. Kazakoff1, Conrad Leonard1, Conrad Leonard2, Scott Wood1, Scott Wood2, Qinying Xu1, Qinying Xu2, 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. Whitehall2, Vicki L. J. Whitehall1, 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. Scarlett4, Christopher J. Scarlett8, 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 Chou11, Angela Chou4, 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. Kench19, James G. Kench4, James G. Kench18, Neil D. Merrett20, Neil D. Merrett18, 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. Gill18, Anthony J. Gill4, David A. Wheeler13, Richard A. Gibbs13, Elizabeth A. Musgrove3, Claudio Bassi, Giampaolo Tortora, Paolo Pederzoli, John V. Pearson2, John V. Pearson1, 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
TL;DR: By parsing the unique classes and subclasses of tumor immune microenvironment (TIME) that exist within a patient’s tumor, the ability to predict and guide immunotherapeutic responsiveness will improve, and new therapeutic targets will be revealed.
Abstract: The clinical successes in immunotherapy have been both astounding and at the same time unsatisfactory. Countless patients with varied tumor types have seen pronounced clinical response with immunotherapeutic intervention; however, many more patients have experienced minimal or no clinical benefit when provided the same treatment. As technology has advanced, so has the understanding of the complexity and diversity of the immune context of the tumor microenvironment and its influence on response to therapy. It has been possible to identify different subclasses of immune environment that have an influence on tumor initiation and response and therapy; by parsing the unique classes and subclasses of tumor immune microenvironment (TIME) that exist within a patient's tumor, the ability to predict and guide immunotherapeutic responsiveness will improve, and new therapeutic targets will be revealed.

2,920 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 Miller1, David Miller4, 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 Holmes3, Oliver Holmes1, Chin4, Matthew J. Anderson1, Stephen H. Kazakoff3, Stephen H. Kazakoff1, Conrad Leonard1, Conrad Leonard3, Felicity Newell1, Nicola Waddell1, Scott Wood1, Scott Wood3, Qinying Xu1, Qinying Xu3, 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. 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. 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 Hofmann2, Oliver Hofmann9, R. L. Sutherland4, David A. Wheeler5, Anthony J. Gill15, Anthony J. Gill4, Richard A. Gibbs5, John V. Pearson1, John V. Pearson3, 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: Pembrolizumab was associated with significantly longer overall survival and with a lower rate of treatment‐related adverse events than chemotherapy as second‐line therapy for platinum‐refractory advanced urothelial carcinoma.
Abstract: BackgroundPatients with advanced urothelial carcinoma that progresses after platinum-based chemotherapy have a poor prognosis and limited treatment options. MethodsIn this open-label, international, phase 3 trial, we randomly assigned 542 patients with advanced urothelial cancer that recurred or progressed after platinum-based chemotherapy to receive pembrolizumab (a highly selective, humanized monoclonal IgG4κ isotype antibody against programmed death 1 [PD-1]) at a dose of 200 mg every 3 weeks or the investigator’s choice of chemotherapy with paclitaxel, docetaxel, or vinflunine. The coprimary end points were overall survival and progression-free survival, which were assessed among all patients and among patients who had a tumor PD-1 ligand (PD-L1) combined positive score (the percentage of PD-L1–expressing tumor and infiltrating immune cells relative to the total number of tumor cells) of 10% or more. ResultsThe median overall survival in the total population was 10.3 months (95% confidence interval [C...

2,362 citations

Journal ArticleDOI
Robert M. Samstein1, Chung-Han Lee1, Chung-Han Lee2, Alexander N. Shoushtari1, Alexander N. Shoushtari2, Matthew D. Hellmann2, Matthew D. Hellmann1, Ronglai Shen1, Yelena Y. Janjigian1, Yelena Y. Janjigian2, David Barron1, Ahmet Zehir1, Emmet Jordan1, Antonio Omuro1, Thomas Kaley1, Sviatoslav M. Kendall1, Robert J. Motzer2, Robert J. Motzer1, A. Ari Hakimi1, Martin H. Voss1, Martin H. Voss2, Paul Russo1, Jonathan E. Rosenberg1, Jonathan E. Rosenberg2, Gopa Iyer1, Gopa Iyer2, Bernard H. Bochner1, Dean F. Bajorin1, Dean F. Bajorin2, Hikmat Al-Ahmadie1, Jamie E. Chaft1, Jamie E. Chaft2, Charles M. Rudin1, Charles M. Rudin2, Gregory J. Riely2, Gregory J. Riely1, Shrujal S. Baxi2, Shrujal S. Baxi1, Alan L. Ho1, Alan L. Ho2, Richard J. Wong1, David G. Pfister1, David G. Pfister2, 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'Angelo2, Sandra P. D'Angelo1, Leonard B. Saltz1, Leonard B. Saltz2, Zsofia K. Stadler1, Zsofia K. Stadler2, Howard I. Scher2, Howard I. Scher1, José Baselga1, José Baselga2, Pedram Razavi1, Pedram Razavi2, Christopher A. Klebanoff1, Christopher A. Klebanoff2, Rona Yaeger2, Rona Yaeger1, Neil H. Segal1, Neil H. Segal2, 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