Network


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

Hotspot


Dive into the research topics where Takafumi N. Yamaguchi is active.

Publication


Featured researches published by Takafumi N. Yamaguchi.


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 Methods | 2015

Combining tumor genome simulation with crowdsourcing to benchmark somatic single-nucleotide-variant detection

Adam D. Ewing; Kathleen E. Houlahan; Yin Hu; Kyle Ellrott; Cristian Caloian; Takafumi N. Yamaguchi; J Christopher Bare; Christine P'ng; Daryl Waggott; Veronica Y. Sabelnykova; Michael R. Kellen; Thea Norman; David Haussler; Stephen H. Friend; Gustavo Stolovitzky; Adam A. Margolin; Joshua M. Stuart; Paul C. Boutros

The detection of somatic mutations from cancer genome sequences is key to understanding the genetic basis of disease progression, patient survival and response to therapy. Benchmarking is needed for tool assessment and improvement but is complicated by a lack of gold standards, by extensive resource requirements and by difficulties in sharing personal genomic information. To resolve these issues, we launched the ICGC-TCGA DREAM Somatic Mutation Calling Challenge, a crowdsourced benchmark of somatic mutation detection algorithms. Here we report the BAMSurgeon tool for simulating cancer genomes and the results of 248 analyses of three in silico tumors created with it. Different algorithms exhibit characteristic error profiles, and, intriguingly, false positives show a trinucleotide profile very similar to one found in human tumors. Although the three simulated tumors differ in sequence contamination (deviation from normal cell sequence) and in subclonality, an ensemble of pipelines outperforms the best individual pipeline in all cases. BAMSurgeon is available at https://github.com/adamewing/bamsurgeon/.


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.


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.


Nature Genetics | 2017

TMPRSS2–ERG fusion co-opts master transcription factors and activates NOTCH signaling in primary prostate cancer

Ken Kron; Alexander Murison; Stanley Zhou; Vincent Huang; Takafumi N. Yamaguchi; Yu Jia Shiah; Michael Fraser; Theodorus H. van der Kwast; Paul C. Boutros; Robert G. Bristow; Mathieu Lupien

TMPRSS2–ERG (T2E) structural rearrangements typify ∼50% of prostate tumors and result in overexpression of the ERG transcription factor. Using chromatin, genomic and expression data, we show distinct cis-regulatory landscapes between T2E-positive and non-T2E primary prostate tumors, which include clusters of regulatory elements (COREs). This difference is mediated by ERG co-option of HOXB13 and FOXA1, implementing a T2E-specific transcriptional profile. We also report a T2E-specific CORE on the structurally rearranged ERG locus arising from spreading of the TMPRSS2 locus pre-existing CORE, assisting in its overexpression. Finally, we show that the T2E-specific cis-regulatory landscape underlies a vulnerability against the NOTCH pathway. Indeed, NOTCH pathway inhibition antagonizes the growth and invasion of T2E-positive prostate cancer cells. Taken together, our work shows that overexpressed ERG co-opts master transcription factors to deploy a unique cis-regulatory landscape, inducing a druggable dependency on NOTCH signaling in T2E-positive prostate tumors.


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.


bioRxiv | 2014

A Comprehensive Assessment of Somatic Mutation Calling in Cancer Genomes

Tyler Alioto; Sophia Derdak; Timothy Beck; Paul C. Boutros; Lawrence Bower; Ivo Buchhalter; Matthew Eldridge; Nicholas J. Harding; Lawrence E. Heisler; Eivind Hovig; David T. W. Jones; Andy G. Lynch; Sigve Nakken; Paolo Ribeca; Anne-Sophie Sertier; Jared T. Simpson; Paul T. Spellman; Patrick Tarpey; Laurie Tonon; Daniel Vodák; Takafumi N. Yamaguchi; Sergi Beltran Agullo; Marc Dabad; Robert E. Denroche; Philip Ginsbach; Simon Heath; Emanuele Raineri; Charlotte L Anderson; Benedikt Brors; Ruben M. Drews

The emergence of next generation DNA sequencing technology is enabling high-resolution cancer genome analysis. Large-scale projects like the International Cancer Genome Consortium (ICGC) are systematically scanning cancer genomes to identify recurrent somatic mutations. Second generation DNA sequencing, however, is still an evolving technology and procedures, both experimental and analytical, are constantly changing. Thus the research community is still defining a set of best practices for cancer genome data analysis, with no single protocol emerging to fulfil this role. Here we describe an extensive benchmark exercise to identify and resolve issues of somatic mutation calling. Whole genome sequence datasets comprising tumor-normal pairs from two different types of cancer, chronic lymphocytic leukaemia and medulloblastoma, were shared within the ICGC and submissions of somatic mutation calls were compared to verified mutations and to each other. Varying strategies to call mutations, incomplete awareness of sources of artefacts, and even lack of agreement on what constitutes an artefact or real mutation manifested in widely varying mutation call rates and somewhat low concordance among submissions. We conclude that somatic mutation calling remains an unsolved problem. However, we have identified many issues that are easy to remedy that are presented here. Our study highlights critical issues that need to be addressed before this valuable technology can be routinely used to inform clinical decision-making. Abbreviations and Definitions SSM Somatic Single-base Mutations or Simple Somatic Mutations, refers to a somatic single base change SIM Somatic Insertion/deletion Mutation CNV Copy Number Variant SV Structural Variant SNP Single Nucleotide Polymorphisms, refers to a single base variable position in the germline with a frequency of > 1% in the general population CLL Chronic Lymphocytic Leukaemia MB Medulloblastoma ICGC International Cancer Genome Consortium BM Benchmark aligner = mapper, these terms are used interchangeably


BMC Bioinformatics | 2018

Germline contamination and leakage in whole genome somatic single nucleotide variant detection

Dorota H Sendorek; Cristian Caloian; Kyle Ellrott; J Christopher Bare; Takafumi N. Yamaguchi; Adam D. Ewing; Kathleen E. Houlahan; Thea Norman; Adam A. Margolin; Joshua M. Stuart; Paul C. Boutros

BackgroundThe clinical sequencing of cancer genomes to personalize therapy is becoming routine across the world. However, concerns over patient re-identification from these data lead to questions about how tightly access should be controlled. It is not thought to be possible to re-identify patients from somatic variant data. However, somatic variant detection pipelines can mistakenly identify germline variants as somatic ones, a process called “germline leakage”. The rate of germline leakage across different somatic variant detection pipelines is not well-understood, and it is uncertain whether or not somatic variant calls should be considered re-identifiable. To fill this gap, we quantified germline leakage across 259 sets of whole-genome somatic single nucleotide variant (SNVs) predictions made by 21 teams as part of the ICGC-TCGA DREAM Somatic Mutation Calling Challenge.ResultsThe median somatic SNV prediction set contained 4325 somatic SNVs and leaked one germline polymorphism. The level of germline leakage was inversely correlated with somatic SNV prediction accuracy and positively correlated with the amount of infiltrating normal cells. The specific germline variants leaked differed by tumour and algorithm. To aid in quantitation and correction of leakage, we created a tool, called GermlineFilter, for use in public-facing somatic SNV databases.ConclusionsThe potential for patient re-identification from leaked germline variants in somatic SNV predictions has led to divergent open data access policies, based on different assessments of the risks. Indeed, a single, well-publicized re-identification event could reshape public perceptions of the values of genomic data sharing. We find that modern somatic SNV prediction pipelines have low germline-leakage rates, which can be further reduced, especially for cloud-sharing, using pre-filtering software.


bioRxiv | 2018

Accurate Reference-Free Somatic Variant-Calling by Integrating Genomic, Sequencing and Population Data

Ren X. Sun; Christopher M Lalansingh; Shadrielle Melijah G. Espiritu; Cindy Q. Yao; Takafumi N. Yamaguchi; Stephenie D. Prokopec; Lesia Szyca; Kathleen E. Houlahan; Lawrence E. Heisler; Morgan Black; Constance H. Li; John W. Barrett; Anthony Charles Nichols; Paul C. Boutros

The detection of somatic single nucleotide variants (SNVs) is critical in both research and clinical applications. Studies of human cancer typically use matched normal (reference) samples from a distant tissue to increase SNV prediction accuracy. This process both doubles sequencing costs and poses challenges when reference samples are not readily available, such as for many cell-lines. To address these challenges, we created S22S: an approach for the prediction of somatic mutations without need for matched reference tissue. S22S takes underlying sequence data, augments them with genomic background context and population frequency information, and classifies SNVs as somatic or non-somatic. We validated S22S using primary tumor/normal pairs from four tumor types, spanning two different sequencing technologies. S22S robustly identifies somatic SNVs, with the area under the precision recall curve reaching 0.97 in kidney clear cell carcinoma, comparable to the best tumor/normal analysis pipelines. S22S is freely available at http://labs.oicr.on.ca/Boutros-lab/software/s22s.


BMC Bioinformatics | 2018

Valection: design optimization for validation and verification studies

Christopher I Cooper; Delia Yao; Dorota H Sendorek; Takafumi N. Yamaguchi; Christine Ng; Kathleen E. Houlahan; Cristian Caloian; Michael Fraser; Smc-Dna Challenge Participants; Kyle Ellrott; Adam A. Margolin; Robert G. Bristow; Joshua M. Stuart; Paul Boutros

BackgroundPlatform-specific error profiles necessitate confirmatory studies where predictions made on data generated using one technology are additionally verified by processing the same samples on an orthogonal technology. However, verifying all predictions can be costly and redundant, and testing a subset of findings is often used to estimate the true error profile.ResultsTo determine how to create subsets of predictions for validation that maximize accuracy of global error profile inference, we developed Valection, a software program that implements multiple strategies for the selection of verification candidates. We evaluated these selection strategies on one simulated and two experimental datasets.ConclusionsValection is implemented in multiple programming languages, available at: http://labs.oicr.on.ca/boutros-lab/software/valection

Collaboration


Dive into the Takafumi N. Yamaguchi's collaboration.

Top Co-Authors

Avatar

Paul C. Boutros

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
Top Co-Authors

Avatar

Michael Fraser

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Vincent Huang

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Julie Livingstone

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emilie Lalonde

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

Kathleen E. Houlahan

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Shadrielle Melijah G. Espiritu

Ontario Institute for Cancer Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge