Network


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

Hotspot


Dive into the research topics where Michelle Chan-Seng-Yue is active.

Publication


Featured researches published by Michelle Chan-Seng-Yue.


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 Genetics | 2014

Hotspot activating PRKD1 somatic mutations in polymorphous low-grade adenocarcinomas of the salivary glands

Ilan Weinreb; Salvatore Piscuoglio; Luciano G. Martelotto; Daryl Waggott; Charlotte K.Y. Ng; Bayardo Perez-Ordonez; Nicholas J. Harding; Javier A. Alfaro; Kenneth C. Chu; Agnes Viale; Nicola Fusco; Arnaud Da Cruz Paula; Caterina Marchiò; Rita A. Sakr; Raymond S. Lim; Lester D R Thompson; Simion I. Chiosea; Raja R. Seethala; Alena Skalova; Edward B. Stelow; Isabel Fonseca; Adel Assaad; Christine How; Jianxin Wang; Richard de Borja; Michelle Chan-Seng-Yue; Christopher J. Howlett; Anthony C. Nichols; Y Hannah Wen; Nora Katabi

Polymorphous low-grade adenocarcinoma (PLGA) is the second most frequent type of malignant tumor of the minor salivary glands. We identified PRKD1 hotspot mutations encoding p.Glu710Asp in 72.9% of PLGAs but not in other salivary gland tumors. Functional studies demonstrated that this kinase-activating alteration likely constitutes a driver of PLGA.


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.


International Scholarly Research Notices | 2012

A Pilot Study Comparing HPV-Positive and HPV-Negative Head and Neck Squamous Cell Carcinomas by Whole Exome Sequencing

Anthony C. Nichols; Michelle Chan-Seng-Yue; John Yoo; Wei Xu; Sandeep Dhaliwal; John Basmaji; Christopher C. T. Szeto; Samuel A. Dowthwaite; Biljana Todorovic; Maud H. W. Starmans; Philippe Lambin; David A. Palma; Kevin Fung; Jason H. Franklin; Bret Wehrli; Keith Kwan; James Koropatnick; Joe S. Mymryk; Paul C. Boutros; John W. Barrett

Background. Next-generation sequencing of cancers has identified important therapeutic targets and biomarkers. The goal of this pilot study was to compare the genetic changes in a human papillomavirus- (HPV-)positive and an HPV-negative head and neck tumor. Methods. DNA was extracted from the blood and primary tumor of a patient with an HPV-positive tonsillar cancer and those of a patient with an HPV-negative oral tongue tumor. Exome enrichment was performed using the Agilent SureSelect All Exon Kit, followed by sequencing on the ABI SOLiD platform. Results. Exome sequencing revealed slightly more mutations in the HPV-negative tumor (73) in contrast to the HPV-positive tumor (58). Multiple mutations were noted in zinc finger genes (ZNF3, 10, 229, 470, 543, 616, 664, 638, 716, and 799) and mucin genes (MUC4, 6, 12, and 16). Mutations were noted in MUC12 in both tumors. Conclusions. HPV-positive HNSCC is distinct from HPV-negative disease in terms of evidence of viral infection, p16 status, and frequency of mutations. Next-generation sequencing has the potential to identify novel therapeutic targets and biomarkers in HNSCC.


Cancer Research | 2013

MYC Phosphorylation at Novel Regulatory Regions Suppresses Transforming Activity

Amanda R. Wasylishen; Michelle Chan-Seng-Yue; Christina Bros; Dharmendra Dingar; William B. Tu; Manpreet Kalkat; Pak Kei Chan; Peter Mullen; Ling Huang; Natalie Meyer; Brian Raught; Paul C. Boutros; Linda Z. Penn

Despite its central role in human cancer, MYC deregulation is insufficient by itself to transform cells. Because inherent mechanisms of neoplastic control prevent precancerous lesions from becoming fully malignant, identifying transforming alleles of MYC that bypass such controls may provide fundamental insights into tumorigenesis. To date, the only activated allele of MYC known is T58A, the study of which led to identification of the tumor suppressor FBXW7 and its regulator USP28 as a novel therapeutic target. In this study, we screened a panel of MYC phosphorylation mutants for their ability to promote anchorage-independent colony growth of human MCF10A mammary epithelial cells, identifying S71A/S81A and T343A/S344A/S347A/S348A as more potent oncogenic mutants compared with wild-type (WT) MYC. The increased cell-transforming activity of these mutants was confirmed in SH-EP neuroblastoma cells and in three-dimensional MCF10A acini. Mechanistic investigations initiated by a genome-wide mRNA expression analysis of MCF10A acini identified 158 genes regulated by the mutant MYC alleles, compared with only 112 genes regulated by both WT and mutant alleles. Transcriptional gain-of-function was a common feature of the mutant alleles, with many additional genes uniquely dysregulated by individual mutant. Our work identifies novel sites of negative regulation in MYC and thus new sites for its therapeutic attack.


Cancer Letters | 2016

Candidate DNA repair susceptibility genes identified by exome sequencing in high-risk pancreatic cancer

Alyssa L. Smith; Najmeh Alirezaie; Ashton A. Connor; Michelle Chan-Seng-Yue; Robert Grant; Iris Selander; Claire Bascuñana; Ayelet Borgida; Anita Hall; Thomas Whelan; Spring Holter; Treasa McPherson; Sean P. Cleary; Gloria M. Petersen; Atilla Omeroglu; Emmanouil Saloustros; John McPherson; Lincoln Stein; William D. Foulkes; Jacek Majewski; Steven Gallinger; George Zogopoulos

The genetic basis underlying the majority of hereditary pancreatic adenocarcinoma (PC) is unknown. Since DNA repair genes are widely implicated in gastrointestinal malignancies, including PC, we hypothesized that there are novel DNA repair PC susceptibility genes. As germline DNA repair gene mutations may lead to PC subtypes with selective therapeutic responses, we also hypothesized that there is an overall survival (OS) difference in mutation carriers versus non-carriers. We therefore interrogated the germline exomes of 109 high-risk PC cases for rare protein-truncating variants (PTVs) in 513 putative DNA repair genes. We identified PTVs in 41 novel genes among 36 kindred. Additional genetic evidence for causality was obtained for 17 genes, with FAN1, NEK1 and RHNO1 emerging as the strongest candidates. An OS difference was observed for carriers versus non-carriers of PTVs with early stage (≤IIB) disease. This adverse survival trend in carriers with early stage disease was also observed in an independent series of 130 PC cases. We identified candidate DNA repair PC susceptibility genes and suggest that carriers of a germline PTV in a DNA repair gene with early stage disease have worse survival.


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.


Epigenetics & Chromatin | 2016

Hypoxia increases genome-wide bivalent epigenetic marking by specific gain of H3K27me3

Peggy Prickaerts; Michiel E. Adriaens; Twan van den Beucken; Elizabeth Koch; Ludwig Dubois; V.E.H. Dahlmans; Caroline Gits; Chris T. Evelo; Michelle Chan-Seng-Yue; Bradly G. Wouters; Jan Willem Voncken

BackgroundTrimethylation at histone H3 lysine 4 (H3K4me3) and lysine 27 (H3K27me3) controls gene activity during development and differentiation. Whether H3K4me3 and H3K27me3 changes dynamically in response to altered microenvironmental conditions, including low-oxygen conditions commonly present in solid tumors, is relatively unknown. Demethylation of H3K4me3 and H3K27me3 is mediated by oxygen and 2-oxoglutarate dioxygenases enzymes, suggesting that oxygen deprivation (hypoxia) may influence histone trimethylation. Using the MCF7 breast epithelial adenocarcinoma cell model, we have determined the relationship between epigenomic and transcriptomic reprogramming as a function of fluctuating oxygen tension.ResultsWe find that in MCF7, H3K4me3 and H3K27me3 marks rapidly increase at specific locations throughout the genome and are largely reversed upon reoxygenation. Whereas dynamic changes are relatively highest for H3K27me3 marking under hypoxic conditions, H3K4me3 occupation is identified as the defining epigenetic marker of transcriptional control. In agreement with the global increase of H3K27 trimethylation, we provide direct evidence that the histone H3K27me3 demethylase KDM6B/JMJD3 is inactivated by limited oxygen. In situ immunohistochemical analysis confirms a marked rise of histone trimethylation in hypoxic tumor areas. Acquisition of H3K27me3 at H3K4me3-marked loci results in a striking increase in “bivalent” epigenetic marking. Hypoxia-induced bivalency substantially overlaps with embryonal stem cell-associated genic bivalency and is retained at numerous loci upon reoxygenation. Transcriptional activity is selectively and progressively dampened at bivalently marked loci upon repeated exposure to hypoxia, indicating that this subset of genes uniquely maintains the potential for epigenetic regulation by KDM activity.ConclusionsThese data suggest that dynamic regulation of the epigenetic state within the tumor environment may have important consequences for tumor plasticity and biology.

Collaboration


Dive into the Michelle Chan-Seng-Yue's collaboration.

Top Co-Authors

Avatar

Paul C. Boutros

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

Maud H. W. Starmans

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Jianxin Wang

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

Nicholas J. Harding

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

Taryne Chong

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Emilie Lalonde

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Jeremy Johns

Ontario Institute for Cancer Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge