Network


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

Hotspot


Dive into the research topics where Robert E. Denroche is active.

Publication


Featured researches published by Robert E. Denroche.


Nature Genetics | 2015

Spatial genomic heterogeneity within localized, multifocal prostate cancer

Paul C. Boutros; Michael Fraser; Nicholas J. Harding; Richard de Borja; Dominique Trudel; Emilie Lalonde; Alice Meng; Pablo H. Hennings-Yeomans; Andrew McPherson; Veronica Y. Sabelnykova; Amin Zia; Natalie S. Fox; Julie Livingstone; Yu Jia Shiah; Jianxin Wang; Timothy Beck; Cherry Have; Taryne Chong; Michelle Sam; Jeremy Johns; Lee Timms; Nicholas Buchner; Ada Wong; John D. Watson; Trent T. Simmons; Christine P'ng; Gaetano Zafarana; Francis Nguyen; Xuemei Luo; Kenneth C. Chu

Herein we provide a detailed molecular analysis of the spatial heterogeneity of clinically localized, multifocal prostate cancer to delineate new oncogenes or tumor suppressors. We initially determined the copy number aberration (CNA) profiles of 74 patients with index tumors of Gleason score 7. Of these, 5 patients were subjected to whole-genome sequencing using DNA quantities achievable in diagnostic biopsies, with detailed spatial sampling of 23 distinct tumor regions to assess intraprostatic heterogeneity in focal genomics. Multifocal tumors are highly heterogeneous for single-nucleotide variants (SNVs), CNAs and genomic rearrangements. We identified and validated a new recurrent amplification of MYCL, which is associated with TP53 deletion and unique profiles of DNA damage and transcriptional dysregulation. Moreover, we demonstrate divergent tumor evolution in multifocal cancer and, in some cases, tumors of independent clonal origin. These data represent the first systematic relation of intraprostatic genomic heterogeneity to predicted clinical outcome and inform the development of novel biomarkers that reflect individual prognosis.


Nature | 2016

A renewed model of pancreatic cancer evolution based on genomic rearrangement patterns.

Faiyaz Notta; Michelle Chan-Seng-Yue; Mathieu Lemire; Yilong Li; Gavin Wilson; Ashton A. Connor; Robert E. Denroche; Sheng Ben Liang; Andrew M.K. Brown; Jaeseung C. Kim; Tao Wang; Jared T. Simpson; Timothy Beck; Ayelet Borgida; Nicholas Buchner; Dianne Chadwick; Sara Hafezi-Bakhtiari; John E. Dick; Lawrence E. Heisler; Michael A. Hollingsworth; Emin Ibrahimov; Gun Ho Jang; Jeremy Johns; Lars G T Jorgensen; Calvin Law; Olga Ludkovski; Ilinca Lungu; Karen Ng; Danielle Pasternack; Gloria M. Petersen

Pancreatic cancer, a highly aggressive tumour type with uniformly poor prognosis, exemplifies the classically held view of stepwise cancer development. The current model of tumorigenesis, based on analyses of precursor lesions, termed pancreatic intraepithelial neoplasm (PanINs) lesions, makes two predictions: first, that pancreatic cancer develops through a particular sequence of genetic alterations (KRAS, followed by CDKN2A, then TP53 and SMAD4); and second, that the evolutionary trajectory of pancreatic cancer progression is gradual because each alteration is acquired independently. A shortcoming of this model is that clonally expanded precursor lesions do not always belong to the tumour lineage, indicating that the evolutionary trajectory of the tumour lineage and precursor lesions can be divergent. This prevailing model of tumorigenesis has contributed to the clinical notion that pancreatic cancer evolves slowly and presents at a late stage. However, the propensity for this disease to rapidly metastasize and the inability to improve patient outcomes, despite efforts aimed at early detection, suggest that pancreatic cancer progression is not gradual. Here, using newly developed informatics tools, we tracked changes in DNA copy number and their associated rearrangements in tumour-enriched genomes and found that pancreatic cancer tumorigenesis is neither gradual nor follows the accepted mutation order. Two-thirds of tumours harbour complex rearrangement patterns associated with mitotic errors, consistent with punctuated equilibrium as the principal evolutionary trajectory. In a subset of cases, the consequence of such errors is the simultaneous, rather than sequential, knockout of canonical preneoplastic genetic drivers that are likely to set-off invasive cancer growth. These findings challenge the current progression model of pancreatic cancer and provide insights into the mutational processes that give rise to these aggressive tumours.


Nature Communications | 2015

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

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

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


Nature | 2017

Genomic hallmarks of localized, non-indolent prostate cancer

Michael Fraser; Veronica Y. Sabelnykova; Takafumi N. Yamaguchi; Lawrence E. Heisler; Julie Livingstone; Vincent Huang; Yu Jia Shiah; Fouad Yousif; Xihui Lin; Andre P. Masella; Natalie S. Fox; Michael Xie; Stephenie D. Prokopec; Alejandro Berlin; Emilie Lalonde; Musaddeque Ahmed; Dominique Trudel; Xuemei Luo; Timothy Beck; Alice Meng; Junyan Zhang; Alister D'Costa; Robert E. Denroche; Haiying Kong; Shadrielle Melijah G. Espiritu; Melvin Lee Kiang Chua; Ada Wong; Taryne Chong; Michelle Sam; Jeremy Johns

Prostate tumours are highly variable in their response to therapies, but clinically available prognostic factors can explain only a fraction of this heterogeneity. Here we analysed 200 whole-genome sequences and 277 additional whole-exome sequences from localized, non-indolent prostate tumours with similar clinical risk profiles, and carried out RNA and methylation analyses in a subset. These tumours had a paucity of clinically actionable single nucleotide variants, unlike those seen in metastatic disease. Rather, a significant proportion of tumours harboured recurrent non-coding aberrations, large-scale genomic rearrangements, and alterations in which an inversion repressed transcription within its boundaries. Local hypermutation events were frequent, and correlated with specific genomic profiles. Numerous molecular aberrations were prognostic for disease recurrence, including several DNA methylation events, and a signature comprised of these aberrations outperformed well-described prognostic biomarkers. We suggest that intensified treatment of genomically aggressive localized prostate cancer may improve cure rates.


JAMA Oncology | 2017

Association of distinct mutational signatures with correlates of increased immune activity in pancreatic ductal adenocarcinoma

Ashton A. Connor; Robert E. Denroche; Gun Ho Jang; Lee Timms; Sangeetha N. Kalimuthu; Iris Selander; Treasa McPherson; Gavin Wilson; Michelle Chan-Seng-Yue; Ivan Borozan; Vincent Ferretti; Robert C. Grant; Ilinca Lungu; Eithne Costello; William Greenhalf; Daniel H. Palmer; Paula Ghaneh; John P. Neoptolemos; Markus W. Büchler; Gloria M. Petersen; Sarah P. Thayer; Michael A. Hollingsworth; Alana Sherker; Daniel Durocher; Neesha C. Dhani; David W. Hedley; Stefano Serra; Aaron Pollett; Michael H. Roehrl; Prashant Bavi

Importance Outcomes for patients with pancreatic ductal adenocarcinoma (PDAC) remain poor. Advances in next-generation sequencing provide a route to therapeutic approaches, and integrating DNA and RNA analysis with clinicopathologic data may be a crucial step toward personalized treatment strategies for this disease. Objective To classify PDAC according to distinct mutational processes, and explore their clinical significance. Design, Setting, and Participants We performed a retrospective cohort study of resected PDAC, using cases collected between 2008 and 2015 as part of the International Cancer Genome Consortium. The discovery cohort comprised 160 PDAC cases from 154 patients (148 primary; 12 metastases) that underwent tumor enrichment prior to whole-genome and RNA sequencing. The replication cohort comprised 95 primary PDAC cases that underwent whole-genome sequencing and expression microarray on bulk biospecimens. Main Outcomes and Measures Somatic mutations accumulate from sequence-specific processes creating signatures detectable by DNA sequencing. Using nonnegative matrix factorization, we measured the contribution of each signature to carcinogenesis, and used hierarchical clustering to subtype each cohort. We examined expression of antitumor immunity genes across subtypes to uncover biomarkers predictive of response to systemic therapies. Results The discovery cohort was 53% male (n = 79) and had a median age of 67 (interquartile range, 58-74) years. The replication cohort was 50% male (n = 48) and had a median age of 68 (interquartile range, 60-75) years. Five predominant mutational subtypes were identified that clustered PDAC into 4 major subtypes: age related, double-strand break repair, mismatch repair, and 1 with unknown etiology (signature 8). These were replicated and validated. Signatures were faithfully propagated from primaries to matched metastases, implying their stability during carcinogenesis. Twelve of 27 (45%) double-strand break repair cases lacked germline or somatic events in canonical homologous recombination genes—BRCA1, BRCA2, or PALB2. Double-strand break repair and mismatch repair subtypes were associated with increased expression of antitumor immunity, including activation of CD8-positive T lymphocytes (GZMA and PRF1) and overexpression of regulatory molecules (cytotoxic T-lymphocyte antigen 4, programmed cell death 1, and indolamine 2,3-dioxygenase 1), corresponding to higher frequency of somatic mutations and tumor-specific neoantigens. Conclusions and Relevance Signature-based subtyping may guide personalized therapy of PDAC in the context of biomarker-driven prospective trials.


Carcinogenesis | 2016

Fine-mapping of chromosome 5p15.33 based on a targeted deep sequencing and high density genotyping identifies novel lung cancer susceptibility loci

Linda Kachuri; Christopher I. Amos; James D. McKay; Mattias Johansson; Paolo Vineis; H. Bas Bueno-de-Mesquita; Marie Christine Boutron-Ruault; Mikael Johansson; J. Ramón Quirós; Sabina Sieri; Ruth C. Travis; Elisabete Weiderpass; Loic Le Marchand; Brian E. Henderson; Lynne R. Wilkens; Gary E. Goodman; Chu Chen; Jennifer A. Doherty; David C. Christiani; Yongyue Wei; Li Su; Shelley S. Tworoger; Xuehong Zhang; Peter Kraft; David Zaridze; John K. Field; Michael W. Marcus; Michael P.A. Davies; Russell Hyde; Neil E. Caporaso

Chromosome 5p15.33 has been identified as a lung cancer susceptibility locus, however the underlying causal mechanisms were not fully elucidated. Previous fine-mapping studies of this locus have relied on imputation or investigated a small number of known, common variants. This study represents a significant advance over previous research by investigating a large number of novel, rare variants, as well as their underlying mechanisms through telomere length. Variants for this fine-mapping study were identified through a targeted deep sequencing (average depth of coverage greater than 4000×) of 576 individuals. Subsequently, 4652 SNPs, including 1108 novel SNPs, were genotyped in 5164 cases and 5716 controls of European ancestry. After adjusting for known risk loci, rs2736100 and rs401681, we identified a new, independent lung cancer susceptibility variant in LPCAT1: rs139852726 (OR = 0.46, P = 4.73×10(-9)), and three new adenocarcinoma risk variants in TERT: rs61748181 (OR = 0.53, P = 2.64×10(-6)), rs112290073 (OR = 1.85, P = 1.27×10(-5)), rs138895564 (OR = 2.16, P = 2.06×10(-5); among young cases, OR = 3.77, P = 8.41×10(-4)). In addition, we found that rs139852726 (P = 1.44×10(-3)) was associated with telomere length in a sample of 922 healthy individuals. The gene-based SKAT-O analysis implicated TERT as the most relevant gene in the 5p15.33 region for adenocarcinoma (P = 7.84×10(-7)) and lung cancer (P = 2.37×10(-5)) risk. In this largest fine-mapping study to investigate a large number of rare and novel variants within 5p15.33, we identified novel lung and adenocarcinoma susceptibility loci with large effects and provided support for the role of telomere length as the potential underlying mechanism.


Cancer Discovery | 2018

Organoid profiling identifies common responders to chemotherapy in pancreatic cancer

Hervé Tiriac; Pascal Belleau; Dannielle D. Engle; Dennis Plenker; Astrid Deschênes; Tim D.D. Somerville; Fieke E.M. Froeling; Richard A. Burkhart; Robert E. Denroche; Gun-Ho Jang; Koji Miyabayashi; C. Megan Young; Hardik Patel; Michelle Ma; Joseph F. LaComb; Randze Lerie D. Palmaira; Ammar A. Javed; Jasmine Huynh; Molly Johnson; Kanika Arora; Nicolas Robine; Minita Shah; Rashesh Sanghvi; Austin Goetz; Cinthya Y. Lowder; Laura Martello; Else Driehuis; Nicolas Lecomte; Gokce Askan; Christine A. Iacobuzio-Donahue

Pancreatic cancer is the most lethal common solid malignancy. Systemic therapies are often ineffective, and predictive biomarkers to guide treatment are urgently needed. We generated a pancreatic cancer patient-derived organoid (PDO) library that recapitulates the mutational spectrum and transcriptional subtypes of primary pancreatic cancer. New driver oncogenes were nominated and transcriptomic analyses revealed unique clusters. PDOs exhibited heterogeneous responses to standard-of-care chemotherapeutics and investigational agents. In a case study manner, we found that PDO therapeutic profiles paralleled patient outcomes and that PDOs enabled longitudinal assessment of chemosensitivity and evaluation of synchronous metastases. We derived organoid-based gene expression signatures of chemosensitivity that predicted improved responses for many patients to chemotherapy in both the adjuvant and advanced disease settings. Finally, we nominated alternative treatment strategies for chemorefractory PDOs using targeted agent therapeutic profiling. We propose that combined molecular and therapeutic profiling of PDOs may predict clinical response and enable prospective therapeutic selection.Significance: New approaches to prioritize treatment strategies are urgently needed to improve survival and quality of life for patients with pancreatic cancer. Combined genomic, transcriptomic, and therapeutic profiling of PDOs can identify molecular and functional subtypes of pancreatic cancer, predict therapeutic responses, and facilitate precision medicine for patients with pancreatic cancer. Cancer Discov; 8(9); 1112-29. ©2018 AACR.See related commentary by Collisson, p. 1062This article is highlighted in the In This Issue feature, p. 1047.


Clinical Cancer Research | 2017

Genomics-Driven Precision Medicine for Advanced Pancreatic Cancer: Early Results from the COMPASS Trial.

Kyaw Lwin Aung; Sandra Fischer; Robert E. Denroche; Gun-Ho Jang; Anna Dodd; Sean Creighton; Bernadette Southwood; Sheng-Ben Liang; Dianne Chadwick; Amy Zhang; Grainne M. O'Kane; Hamzeh Albaba; Shari Moura; Robert C. Grant; Jessica Miller; Faridah Mbabaali; Danielle Pasternack; Ilinca Lungu; John M. S. Bartlett; Sangeet Ghai; Mathieu Lemire; Spring Holter; Ashton A. Connor; Richard A. Moffitt; Jen Jen Yeh; Lee Timms; Paul M. Krzyzanowski; Neesha C. Dhani; David W. Hedley; Faiyaz Notta

Purpose: To perform real-time whole genome sequencing (WGS) and RNA sequencing (RNASeq) of advanced pancreatic ductal adenocarcinoma (PDAC) to identify predictive mutational and transcriptional features for better treatment selection. Experimental Design: Patients with advanced PDAC were prospectively recruited prior to first-line combination chemotherapy. Fresh tumor tissue was acquired by image-guided percutaneous core biopsy for WGS and RNASeq. Laser capture microdissection was performed for all cases. Primary endpoint was feasibility to report WGS results prior to first disease assessment CT scan at 8 weeks. The main secondary endpoint was discovery of patient subsets with predictive mutational and transcriptional signatures. Results: Sixty-three patients underwent a tumor biopsy between December 2015 and June 2017. WGS and RNASeq were successful in 62 (98%) and 60 (95%), respectively. Genomic results were reported at a median of 35 days (range, 19–52 days) from biopsy, meeting the primary feasibility endpoint. Objective responses to first-line chemotherapy were significantly better in patients with the classical PDAC RNA subtype compared with those with the basal-like subtype (P = 0.004). The best progression-free survival was observed in those with classical subtype treated with m-FOLFIRINOX. GATA6 expression in tumor measured by RNA in situ hybridization was found to be a robust surrogate biomarker for differentiating classical and basal-like PDAC subtypes. Potentially actionable genetic alterations were found in 30% of patients. Conclusions: Prospective genomic profiling of advanced PDAC is feasible, and our early data indicate that chemotherapy response differs among patients with different genomic/transcriptomic subtypes. Clin Cancer Res; 24(6); 1344–54. ©2017 AACR.


PLOS ONE | 2014

A Two-Dimensional Pooling Strategy for Rare Variant Detection on Next-Generation Sequencing Platforms

Philip C. Zuzarte; Robert E. Denroche; Gordon Fehringer; Hagit Katzov-Eckert; Rayjean J. Hung; John D. McPherson

We describe a method for pooling and sequencing DNA from a large number of individual samples while preserving information regarding sample identity. DNA from 576 individuals was arranged into four 12 row by 12 column matrices and then pooled by row and by column resulting in 96 total pools with 12 individuals in each pool. Pooling of DNA was carried out in a two-dimensional fashion, such that DNA from each individual is present in exactly one row pool and exactly one column pool. By considering the variants observed in the rows and columns of a matrix we are able to trace rare variants back to the specific individuals that carry them. The pooled DNA samples were enriched over a 250 kb region previously identified by GWAS to significantly predispose individuals to lung cancer. All 96 pools (12 row and 12 column pools from 4 matrices) were barcoded and sequenced on an Illumina HiSeq 2000 instrument with an average depth of coverage greater than 4,000×. Verification based on Ion PGM sequencing confirmed the presence of 91.4% of confidently classified SNVs assayed. In this way, each individual sample is sequenced in multiple pools providing more accurate variant calling than a single pool or a multiplexed approach. This provides a powerful method for rare variant detection in regions of interest at a reduced cost to the researcher.


bioRxiv | 2014

A comprehensive multicenter comparison of whole genome sequencing pipelines using a uniform tumor-normal sample pair

Ivo Buchhalter; Barbara Hutter; Tyler Alioto; Timothy Beck; Paul C. Boutros; Benedikt Brors; Adam Butler; Sasithorn Chotewutmontri; Robert E. Denroche; Sophia Derdak; Nicolle Diessl; Lars Feuerbach; Akihiro Fujimoto; Susanne Gröbner; Marta Gut; Nicholas J. Harding; Michael Heinold; Lawrence E. Heisler; Jonathan Hinton; Natalie Jäger; David Jones; Rolf Kabbe; Andrey Korshunov; John D. McPherson; Andrew Menzies; Hidewaki Nakagawa; Christopher Previti; Keiran Raine; Paolo Ribeca; Sabine Schmidt

As next-generation sequencing becomes a clinical tool, a full understanding of the variables affecting sequencing analysis output is required. Through the International Cancer Genome Consortium (ICGC), we compared sequencing pipelines at five independent centers (CNAG, DKFZ, OICR, RIKEN and WTSI) using a single tumor-blood DNA pair. Analyses by each center and with one standardized algorithm revealed significant discrepancies. Although most pipelines performed well for coding mutations, library preparation methods and sequencing coverage metrics clearly influenced downstream results. PCR-free methods showed reduced GC-bias and more even coverage. Increasing sequencing depth to ∼100x (two- to three-fold higher than current standards) showed a benefit, as long as the tumor:control coverage ratio remained balanced. To become part of routine clinical care, high-throughput sequencing must be globally compatible and comparable. This benchmarking exercise has highlighted several fundamental parameters to consider in this regard, which will allow for better optimization and planning of both basic and translational studies.

Collaboration


Dive into the Robert E. Denroche's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lincoln Stein

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Timothy Beck

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ilinca Lungu

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Lee Timms

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Faiyaz Notta

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Julie M. Wilson

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Lawrence E. Heisler

Ontario Institute for Cancer Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge