Network


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

Hotspot


Dive into the research topics where Lee Timms is active.

Publication


Featured researches published by Lee Timms.


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 | 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.


Nature Communications | 2017

Germline BRCA2 mutations drive prostate cancers with distinct evolutionary trajectories

Renea A. Taylor; Michael Fraser; Julie Livingstone; Shadrielle Melijah G. Espiritu; Heather Thorne; Vincent Huang; Winnie Lo; Yu Jia Shiah; Takafumi N. Yamaguchi; Ania Sliwinski; Sheri Horsburgh; Alice Meng; Lawrence E. Heisler; Nancy Yu; Fouad Yousif; Melissa Papargiris; Mitchell G. Lawrence; Lee Timms; Declan Murphy; Mark Frydenberg; Julia F. Hopkins; Damien Bolton; David Clouston; John D. McPherson; Theodorus van der Kwast; Paul C. Boutros; Gail P. Risbridger; Robert G. Bristow

Germline mutations in the BRCA2 tumour suppressor are associated with both an increased lifetime risk of developing prostate cancer (PCa) and increased risk of aggressive disease. To understand this aggression, here we profile the genomes and methylomes of localized PCa from 14 carriers of deleterious germline BRCA2 mutations (BRCA2-mutant PCa). We show that BRCA2-mutant PCa harbour increased genomic instability and a mutational profile that more closely resembles metastastic than localized disease. BRCA2-mutant PCa shows genomic and epigenomic dysregulation of the MED12L/MED12 axis, which is frequently dysregulated in metastatic castration-resistant prostate cancer (mCRPC). This dysregulation is enriched in BRCA2-mutant PCa harbouring intraductal carcinoma (IDC). Microdissection and sequencing of IDC and juxtaposed adjacent non-IDC invasive carcinoma in 10 patients demonstrates a common ancestor to both histopathologies. Overall we show that localized castration-sensitive BRCA2-mutant tumours are uniquely aggressive, due to de novo aberration in genes usually associated with metastatic disease, justifying aggressive initial treatment.


International Journal of Cancer | 2014

Identification of genes expressed by immune cells of the colon that are regulated by colorectal cancer-associated variants

Vanya Peltekova; Mathieu Lemire; Aamer Mahmood Qazi; Syed H. Zaidi; Quang M. Trinh; Ryszard Bielecki; Marianne Rogers; Lyndsey Hodgson; Mike Wang; David J. A. D'Souza; Sasan Zandi; Taryne Chong; Jennifer Y. Y. Kwan; Krystian Kozak; Richard de Borja; Lee Timms; Jagadish Rangrej; Milica Volar; Michelle Chan-Seng-Yue; Timothy Beck; Colleen Ash; Shawna Lee; Jianxin Wang; Paul C. Boutros; Lincoln Stein; John E. Dick; Robert Gryfe; John D. McPherson; Brent W. Zanke; Aaron Pollett

A locus on human chromosome 11q23 tagged by marker rs3802842 was associated with colorectal cancer (CRC) in a genome‐wide association study; this finding has been replicated in case–control studies worldwide. In order to identify biologic factors at this locus that are related to the etiopathology of CRC, we used microarray‐based target selection methods, coupled to next‐generation sequencing, to study 103 kb at the 11q23 locus. We genotyped 369 putative variants from 1,030 patients with CRC (cases) and 1,061 individuals without CRC (controls) from the Ontario Familial Colorectal Cancer Registry. Two previously uncharacterized genes, COLCA1 and COLCA2, were found to be co‐regulated genes that are transcribed from opposite strands. Expression levels of COLCA1 and COLCA2 transcripts correlate with rs3802842 genotypes. In colon tissues, COLCA1 co‐localizes with crystalloid granules of eosinophils and granular organelles of mast cells, neutrophils, macrophages, dendritic cells and differentiated myeloid‐derived cell lines. COLCA2 is present in the cytoplasm of normal epithelial, immune and other cell lineages, as well as tumor cells. Tissue microarray analysis demonstrates the association of rs3802842 with lymphocyte density in the lamina propria (p = 0.014) and levels of COLCA1 in the lamina propria (p = 0.00016) and COLCA2 (tumor cells, p = 0.0041 and lamina propria, p = 6 × 10–5). In conclusion, genetic, expression and immunohistochemical data implicate COLCA1 and COLCA2 in the pathogenesis of colon cancer. Histologic analyses indicate the involvement of immune pathways.


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

Use of sequenom sample ID plus® SNP genotyping in identification of FFPE tumor samples

Jessica Miller; Nicholas Buchner; Lee Timms; Shirley Tam; Xuemei Luo; Andrew M.K. Brown; Danielle Pasternack; Robert G. Bristow; Michael Fraser; Paul C. Boutros; John D. McPherson

Short tandem repeat (STR) analysis, such as the AmpFlSTR® Identifiler® Plus kit, is a standard, PCR-based human genotyping method used in the field of forensics. Misidentification of cell line and tissue DNA can be costly if not detected early; therefore it is necessary to have quality control measures such as STR profiling in place. A major issue in large-scale research studies involving archival formalin-fixed paraffin embedded (FFPE) tissues is that varying levels of DNA degradation can result in failure to correctly identify samples using STR genotyping. PCR amplification of STRs of several hundred base pairs is not always possible when DNA is degraded. The Sample ID Plus® panel from Sequenom allows for human DNA identification and authentication using SNP genotyping. In comparison to lengthy STR amplicons, this multiplexing PCR assay requires amplification of only 76–139 base pairs, and utilizes 47 SNPs to discriminate between individual samples. In this study, we evaluated both STR and SNP genotyping methods of sample identification, with a focus on paired FFPE tumor/normal DNA samples intended for next-generation sequencing (NGS). The ability to successfully validate the identity of FFPE samples can enable cost savings by reducing rework.


BMC Research Notes | 2015

A cancer cell-line titration series for evaluating somatic classification.

Robert E. Denroche; Laura Mullen; Lee Timms; Timothy Beck; Christina K. Yung; Lincoln Stein; John D. McPherson; Andrew M.K. Brown

BackgroundAccurate detection of somatic single nucleotide variants and small insertions and deletions from DNA sequencing experiments of tumour-normal pairs is a challenging task. Tumour samples are often contaminated with normal cells confounding the available evidence for the somatic variants. Furthermore, tumours are heterogeneous so sub-clonal variants are observed at reduced allele frequencies. We present here a cell-line titration series dataset that can be used to evaluate somatic variant calling pipelines with the goal of reliably calling true somatic mutations at low allele frequencies.ResultsCell-line DNA was mixed with matched normal DNA at 8 different ratios to generate samples with known tumour cellularities, and exome sequenced on Illumina HiSeq to depths of >300×. The data was processed with several different variant calling pipelines and verification experiments were performed to assay >1500 somatic variant candidates using Ion Torrent PGM as an orthogonal technology. By examining the variants called at varying cellularities and depths of coverage, we show that the best performing pipelines are able to maintain a high level of precision at any cellularity. In addition, we estimate the number of true somatic variants undetected as cellularity and coverage decrease.ConclusionsOur cell-line titration series dataset, along with the associated verification results, was effective for this evaluation and will serve as a valuable dataset for future somatic calling algorithm development. The data is available for further analysis at the European Genome-phenome Archive under accession number EGAS00001001016. Data access requires registration through the International Cancer Genome Consortium’s Data Access Compliance Office (ICGC DACO).


Cancer Research | 2016

Abstract 5221: Linking the molecular profile of colorectal tumors to germline genetic and environmental risk factors

Syed H. Zaidi; Catie Grasso; Jasmine Mu; Eve Shinbrot; Marios Giannakis; Charles Connolly; Ivan Borozan; Hermann Brenner; Peter T. Campbell; Andrew T. Chan; Jenny Chang-Claude; Mengmeng Du; Vincent Ferretti; Amy J. French; Charles S. Fuchs; Steven Gallinger; Levi A. Garraway; Andrea Gsur; Marc J. Gunter; Tabitha A. Harrison; Michael Hoffmeister; Li Hsu; Wen-Yi Huang; Jeroen R. Huyghe; Mathieu Lemire; Elaine R. Mardis; John McPherson; Polly A. Newcomb; Lincoln Stein; Wei Sun

The completion of The Cancer Genome Atlas (TCGA) project for colorectal cancer (CRC) has enabled a new, focused phase of sequencing tumor samples for which genome-wide genetic, epidemiological, clinical and lifestyle data have been collected. By combining somatic mutational profiles with these aforementioned data, we may identify and report effective prevention and treatment approaches for a broader population of individuals. The advent of targeted deep sequencing approaches using DNA obtained from formalin fixed paraffin embedded tissues has made possible the genetic characterization of the large numbers of patients needed to make such an effort relevant. As a first step, we describe a custom gene panel designed from large-scale studies for targeted deep sequencing, and its application to over 4,200 CR tumors, collected by the Genetics and Epidemiology of Colorectal Cancer Consortium (GECCO). This study is designed to identify recurrent somatic mutations and copy number alterations (CNAs) for association testing with germline genetic and lifestyle and environmental risk factors for CRC, and thereby identify relevant approaches to impact cancer prevention. The targeted CRC panel includes 205 genes. Genes were primarily selected as significantly mutated genes identified from the Nurses’ Health Study and Health Professional9s Follow-up Study (n = 700), and TCGA (n = 525). We also included 15 genes associated with high penetrance germline mutations and augmented the list to include genes in pathways of somatically altered genes, identified by literature review, from public databases and known to be associated with loss of heterozygosity. For these 205 genes, amplification primers were designed to include all coding regions of transcripts that are listed in the UCSC Genome Browser. For regions with CNAs, the TCGA dataset was analyzed to include regions with greater than or equal to 2 copy focal gains or losses that were found in more than 4 or 3 tumors, respectively. Candidate genes in regions with significant CNAs from the TCGA CRC publication (Nature 2012) were also included. For CNAs, 6 to 12 amplicons were designed for each of the 56 selected regions (32 gains and 24 losses). Additional target regions include: 1) 25 microsatellite loci previously used to identify defective DNA mismatch repair and 212 homoploymer repeats. These include microsatellite loci recommended by the NCI Consensus Panel for identifying MSI; 2) amelogenin (for gender); and 3) Fusobacterium to detect a putative CRC-associated pathogen in tumor biopsies. At the AACR annual meeting, we expect to present results for deep sequencing (∼1,000X) of the first 1,000 CR tumors, including any preliminarily identified pathways and subtypes that may provide the basis for association testing with germline genetic and lifestyle and environmental risk factors needed for inferring better approaches to prevention and treatment of CRC. Citation Format: Syed H. Zaidi, Catie Grasso, Jasmine Mu, Eve Shinbrot, Marios Giannakis, Charles Connolly, Ivan Borozan, Hermann Brenner, Peter Campbell, Andrew Chan, Jenny Chang-Claude, Mengmeng Du, Vincent Ferretti, Amy French, Charles Fuchs, Steven Gallinger, Levi Garraway, Andrea Gsur, Marc Gunter, Tabitha Harrison, Michael Hoffmeister, Li Hsu, Wen-Yi Huang, Jeroen Huyghe, Mathieu Lemire, Elaine Mardis, John McPherson, Polly Newcomb, Lincoln Stein, Wei Sun, Lee Timms, Quang Trinh, David Wheeler, Christina Yung, Niha Zubair, Shuji Ogino, Stephen Thibodeau, Ulrike Peters, Thomas Hudson. Linking the molecular profile of colorectal tumors to germline genetic and environmental risk factors. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 5221.

Collaboration


Dive into the Lee Timms'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

Michelle Sam

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Fouad Yousif

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Michael Fraser

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Paul C. Boutros

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Richard de Borja

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Robert E. Denroche

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Taryne Chong

Ontario Institute for Cancer Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge