Network


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

Hotspot


Dive into the research topics where Adrian Wan is active.

Publication


Featured researches published by Adrian Wan.


Nature | 2015

Dynamics of genomic clones in breast cancer patient xenografts at single-cell resolution.

Peter Eirew; Adi Steif; Jaswinder Khattra; Gavin Ha; Damian Yap; Hossein Farahani; Karen A. Gelmon; Stephen Chia; Colin Mar; Adrian Wan; Emma Laks; Justina Biele; Karey Shumansky; Jamie Rosner; Andrew McPherson; Cydney Nielsen; Andrew Roth; Calvin Lefebvre; Ali Bashashati; Camila P. E. de Souza; Celia Siu; Radhouane Aniba; Jazmine Brimhall; Arusha Oloumi; Tomo Osako; Alejandra Bruna; Jose L. Sandoval; Teresa Ruiz de Algara; Wendy Greenwood; Kaston Leung

Human cancers, including breast cancers, comprise clones differing in mutation content. Clones evolve dynamically in space and time following principles of Darwinian evolution, underpinning important emergent features such as drug resistance and metastasis. Human breast cancer xenoengraftment is used as a means of capturing and studying tumour biology, and breast tumour xenografts are generally assumed to be reasonable models of the originating tumours. However, the consequences and reproducibility of engraftment and propagation on the genomic clonal architecture of tumours have not been systematically examined at single-cell resolution. Here we show, using deep-genome and single-cell sequencing methods, the clonal dynamics of initial engraftment and subsequent serial propagation of primary and metastatic human breast cancers in immunodeficient mice. In all 15 cases examined, clonal selection on engraftment was observed in both primary and metastatic breast tumours, varying in degree from extreme selective engraftment of minor (<5% of starting population) clones to moderate, polyclonal engraftment. Furthermore, ongoing clonal dynamics during serial passaging is a feature of tumours experiencing modest initial selection. Through single-cell sequencing, we show that major mutation clusters estimated from tumour population sequencing relate predictably to the most abundant clonal genotypes, even in clonally complex and rapidly evolving cases. Finally, we show that similar clonal expansion patterns can emerge in independent grafts of the same starting tumour population, indicating that genomic aberrations can be reproducible determinants of evolutionary trajectories. Our results show that measurement of genomically defined clonal population dynamics will be highly informative for functional studies using patient-derived breast cancer xenoengraftment.


Nature Methods | 2014

PyClone: statistical inference of clonal population structure in cancer

Andrew Roth; Jaswinder Khattra; Damian Yap; Adrian Wan; Emma Laks; Justina Biele; Gavin Ha; Samuel Aparicio; Alexandre Bouchard-Côté; Sohrab P. Shah

We introduce PyClone, a statistical model for inference of clonal population structures in cancers. PyClone is a Bayesian clustering method for grouping sets of deeply sequenced somatic mutations into putative clonal clusters while estimating their cellular prevalences and accounting for allelic imbalances introduced by segmental copy-number changes and normal-cell contamination. Single-cell sequencing validation demonstrates PyClones accuracy.


The New England Journal of Medicine | 2012

Recurrent Somatic DICER1 Mutations in Nonepithelial Ovarian Cancers

Alireza Heravi-Moussavi; Michael S. Anglesio; S.-W. Grace Cheng; Janine Senz; Winnie Yang; Leah M Prentice; Anthony P. Fejes; Christine Chow; Alicia A. Tone; Steve E. Kalloger; Nancy Hamel; Andrew Roth; Gavin Ha; Adrian Wan; Sarah Maines-Bandiera; Clara Salamanca; Barbara Pasini; Blaise Clarke; Anna F. Lee; Cheng-Han Lee; Chengquan Zhao; Robert H. Young; Samuel Aparicio; Poul H. Sorensen; Michelle Woo; Niki Boyd; Steven J.M. Jones; Martin Hirst; Marco A. Marra; Blake Gilks

BACKGROUND Germline truncating mutations in DICER1, an endoribonuclease in the RNase III family that is essential for processing microRNAs, have been observed in families with the pleuropulmonary blastoma-family tumor and dysplasia syndrome. Mutation carriers are at risk for nonepithelial ovarian tumors, notably sex cord-stromal tumors. METHODS We sequenced the whole transcriptomes or exomes of 14 nonepithelial ovarian tumors and noted closely clustered mutations in the region of DICER1 encoding the RNase IIIb domain of DICER1 in four samples. We then sequenced this region of DICER1 in additional ovarian tumors and in certain other tumors and queried the effect of the mutations on the enzymatic activity of DICER1 using in vitro RNA cleavage assays. RESULTS DICER1 mutations in the RNase IIIb domain were found in 30 of 102 nonepithelial ovarian tumors (29%), predominantly in Sertoli-Leydig cell tumors (26 of 43, or 60%), including 4 tumors with additional germline DICER1 mutations. These mutations were restricted to codons encoding metal-binding sites within the RNase IIIb catalytic centers, which are critical for microRNA interaction and cleavage, and were somatic in all 16 samples in which germline DNA was available for testing. We also detected mutations in 1 of 14 nonseminomatous testicular germ-cell tumors, in 2 of 5 embryonal rhabdomyosarcomas, and in 1 of 266 epithelial ovarian and endometrial carcinomas. The mutant DICER1 proteins had reduced RNase IIIb activity but retained RNase IIIa activity. CONCLUSIONS Somatic missense mutations affecting the RNase IIIb domain of DICER1 are common in nonepithelial ovarian tumors. These mutations do not obliterate DICER1 function but alter it in specific cell types, a novel mechanism through which perturbation of microRNA processing may be oncogenic. (Funded by the Terry Fox Research Institute and others.).


Nature Genetics | 2016

Divergent modes of clonal spread and intraperitoneal mixing in high-grade serous ovarian cancer

Andrew McPherson; Andrew Roth; Emma Laks; Tehmina Masud; Ali Bashashati; Allen W. Zhang; Gavin Ha; Justina Biele; Damian Yap; Adrian Wan; Leah M Prentice; Jaswinder Khattra; Maia A. Smith; Cydney Nielsen; Sarah C. Mullaly; Steve E. Kalloger; Anthony N. Karnezis; Karey Shumansky; Celia Siu; Jamie Rosner; Hector Li Chan; Julie Ho; Nataliya Melnyk; Janine Senz; Winnie Yang; Richard G. Moore; Andrew J. Mungall; Marco A. Marra; Alexandre Bouchard-Côté; C. Blake Gilks

We performed phylogenetic analysis of high-grade serous ovarian cancers (68 samples from seven patients), identifying constituent clones and quantifying their relative abundances at multiple intraperitoneal sites. Through whole-genome and single-nucleus sequencing, we identified evolutionary features including mutation loss, convergence of the structural genome and temporal activation of mutational processes that patterned clonal progression. We then determined the precise clonal mixtures comprising each tumor sample. The majority of sites were clonally pure or composed of clones from a single phylogenetic clade. However, each patient contained at least one site composed of polyphyletic clones. Five patients exhibited monoclonal and unidirectional seeding from the ovary to intraperitoneal sites, and two patients demonstrated polyclonal spread and reseeding. Our findings indicate that at least two distinct modes of intraperitoneal spread operate in clonal dissemination and highlight the distribution of migratory potential over clonal populations comprising high-grade serous ovarian cancers.


Experimental Hematology | 2008

Linkage of Meis1 leukemogenic activity to multiple downstream effectors including Trib2 and Ccl3

Bob Argiropoulos; Lars Palmqvist; Eric Yung; Florian Kuchenbauer; Michael Heuser; Laura M. Sly; Adrian Wan; Gerald Krystal; R. Keith Humphries

OBJECTIVE MEIS1, a HOX cofactor, collaborates with multiple HOX and NUP98-HOX fusion proteins to accelerate the onset of acute myeloid leukemia (AML) through largely unknown molecular mechanisms. MATERIALS AND METHODS To further resolve these mechanisms, we conducted a structure-function analysis of MEIS1 and gene-expression profiling, in the context of NUP98-HOXD13 (ND13) leukemogenesis. RESULTS We show, in a murine bone marrow transplantation model, that the PBX-interaction domain, the homeodomain, and the C-terminal domain of MEIS1, are all required for leukemogenic collaboration with ND13. In contrast, the N-terminal domain of MEIS1 is dispensable for collaboration with ND13, but is required for Flt3 upregulation, indicating additional roles for MEIS1 in induction of leukemia independent of alterations in Flt3 expression. Gene-expression profiling of a cloned ND13 preleukemic cell line transduced with wild-type or Meis1 mutant forms revealed deregulation of multiple genes, including a set not previously implicated as MEIS1 targets. Chromatin immunoprecipitation revealed the in vivo occupancy of MEIS1 on regulatory sequences of Trib2, Flt3, Dlk1, Ccl3, Ccl4, Pf4, and Rgs1. Furthermore, engineered overexpression of Trib2 complements ND13 to induce AML while Ccl3 potentiates the repopulating ability of ND13. CONCLUSION This study shows that Meis1-induced leukemogenesis with ND13 can occur in the absence of Flt3 upregulation and reveals the existence of other pathways activated by MEIS1 to promote leukemia.


Nature Genetics | 2017

Genomic consequences of aberrant DNA repair mechanisms stratify ovarian cancer histotypes

Yi Kan Wang; Ali Bashashati; Michael S. Anglesio; Dawn R. Cochrane; Diljot Grewal; Gavin Ha; Andrew McPherson; Hugo M. Horlings; Janine Senz; Leah M Prentice; Anthony N. Karnezis; Daniel Lai; Mohamed R Aniba; Allen W. Zhang; Karey Shumansky; Celia Siu; Adrian Wan; Melissa K. McConechy; Hector Li-Chang; Alicia A. Tone; Diane Provencher; Manon de Ladurantaye; Hubert Fleury; Aikou Okamoto; Satoshi Yanagida; Nozomu Yanaihara; Misato Saito; Andrew J. Mungall; Richard G. Moore; Marco A. Marra

We studied the whole-genome point mutation and structural variation patterns of 133 tumors (59 high-grade serous (HGSC), 35 clear cell (CCOC), 29 endometrioid (ENOC), and 10 adult granulosa cell (GCT)) as a substrate for class discovery in ovarian cancer. Ab initio clustering of integrated point mutation and structural variation signatures identified seven subgroups both between and within histotypes. Prevalence of foldback inversions identified a prognostically significant HGSC group associated with inferior survival. This finding was recapitulated in two independent cohorts (n = 576 cases), transcending BRCA1 and BRCA2 mutation and gene expression features of HGSC. CCOC cancers grouped according to APOBEC deamination (26%) and age-related mutational signatures (40%). ENOCs were divided by cases with microsatellite instability (28%), with a distinct mismatch-repair mutation signature. Taken together, our work establishes the potency of the somatic genome, reflective of diverse DNA repair deficiencies, to stratify ovarian cancers into distinct biological strata within the major histotypes.


Nature Methods | 2016

Clonal genotype and population structure inference from single-cell tumor sequencing

Andrew Roth; Andrew McPherson; Emma Laks; Justina Biele; Damian Yap; Adrian Wan; Maia A. Smith; Cydney Nielsen; Jessica N. McAlpine; Samuel Aparicio; Alexandre Bouchard-Côté; Sohrab P. Shah

Single-cell DNA sequencing has great potential to reveal the clonal genotypes and population structure of human cancers. However, single-cell data suffer from missing values and biased allelic counts as well as false genotype measurements owing to the sequencing of multiple cells. We describe the Single Cell Genotyper (https://bitbucket.org/aroth85/scg), an open-source software based on a statistical model coupled with a mean-field variational inference method, which can be used to address these problems and robustly infer clonal genotypes.


Nature Communications | 2017

CLK-dependent exon recognition and conjoined gene formation revealed with a novel small molecule inhibitor

Tyler Funnell; Shinya Tasaki; Arusha Oloumi; Shinsuke Araki; Esther Kong; Damian Yap; Yusuke Nakayama; Christopher S. Hughes; S.-W. Grace Cheng; Hirokazu Tozaki; Misa Iwatani; Satoshi Sasaki; Tomohiro Ohashi; Tohru Miyazaki; Nao Morishita; Daisuke Morishita; Mari Ogasawara-Shimizu; Momoko Ohori; Shoichi Nakao; Masatoshi Karashima; Masaya Sano; Aiko Murai; Toshiyuki Nomura; Noriko Uchiyama; Tomohiro Kawamoto; Ryujiro Hara; Osamu Nakanishi; Karey Shumansky; Jamie Rosner; Adrian Wan

CDC-like kinase phosphorylation of serine/arginine-rich proteins is central to RNA splicing reactions. Yet, the genomic network of CDC-like kinase-dependent RNA processing events remains poorly defined. Here, we explore the connectivity of genomic CDC-like kinase splicing functions by applying graduated, short-exposure, pharmacological CDC-like kinase inhibition using a novel small molecule (T3) with very high potency, selectivity, and cell-based stability. Using RNA-Seq, we define CDC-like kinase-responsive alternative splicing events, the large majority of which monotonically increase or decrease with increasing CDC-like kinase inhibition. We show that distinct RNA-binding motifs are associated with T3 response in skipped exons. Unexpectedly, we observe dose-dependent conjoined gene transcription, which is associated with motif enrichment in the last and second exons of upstream and downstream partners, respectively. siRNA knockdown of CLK2-associated genes significantly increases conjoined gene formation. Collectively, our results reveal an unexpected role for CDC-like kinase in conjoined gene formation, via regulation of 3′-end processing and associated splicing factors.The phosphorylation of serine/arginine-rich proteins by CDC-like kinase is a central regulatory mechanism for RNA splicing reactions. Here, the authors synthesize a novel small molecule CLK inhibitor and map CLK-responsive alternative splicing events and discover an effect on conjoined gene transcription.


Blood | 2011

Ontogeny stage-independent and high-level clonal expansion in vitro of mouse hematopoietic stem cells stimulated by an engineered NUP98-HOX fusion transcription factor

Sanja Sekulovic; Maura Gasparetto; Véronique Lecault; Corinne A. Hoesli; David G. Kent; Patty Rosten; Adrian Wan; Christy Brookes; Carl L. Hansen; James M. Piret; Clayton A. Smith; Connie J. Eaves; R. Keith Humphries

Achieving high-level expansion of hematopoietic stem cells (HSCs) in vitro will have an important clinical impact in addition to enabling elucidation of their regulation. Here, we couple the ability of engineered NUP98-HOXA10hd expression to stimulate > 1000-fold net expansions of murine HSCs in 10-day cultures initiated with bulk lin(-)Sca-1(+)c-kit(+) cells, with strategies to purify fetal and adult HSCs and analyze their expansion clonally. We find that NUP98-HOXA10hd stimulates comparable expansions of HSCs from both sources at ∼ 60% to 90% unit efficiency in cultures initiated with single cells. Clonally expanded HSCs consistently show balanced long-term contributions to the lymphoid and myeloid lineages without evidence of leukemogenic activity. Although effects on fetal and adult HSCs were indistinguishable, NUP98-HOXA10hd-transduced adult HSCs did not thereby gain a competitive advantage in vivo over freshly isolated fetal HSCs. Live-cell image tracking of single transduced HSCs cultured in a microfluidic device indicates that NUP98-HOXA10hd does not affect their proliferation kinetics, and flow cytometry confirmed the phenotype of normal proliferating HSCs and allowed reisolation of large numbers of expanded HSCs at a purity of 25%. These findings point to the effects of NUP98-HOXA10hd on HSCs in vitro being mediated by promoting self-renewal and set the stage for further dissection of this process.


Breast Cancer Research | 2015

A co-culture genome-wide RNAi screen with mammary epithelial cells reveals transmembrane signals required for growth and differentiation

Angela Burleigh; Steven McKinney; Jazmine Brimhall; Damian Yap; Peter Eirew; Steven S.S. Poon; Viola Ng; Adrian Wan; Leah M Prentice; Lois Annab; J. Carl Barrett; Carlos Caldas; Connie J. Eaves; Samuel Aparicio

IntroductionThe extracellular signals regulating mammary epithelial cell growth are of relevance to understanding the pathophysiology of mammary epithelia, yet they remain poorly characterized. In this study, we applied an unbiased approach to understanding the functional role of signalling molecules in several models of normal physiological growth and translated these results to the biological understanding of breast cancer subtypes.MethodsWe developed and utilized a cytogenetically normal clonal line of hTERT immortalized human mammary epithelial cells in a fibroblast-enhanced co-culture assay to conduct a genome-wide small interfering RNA (siRNA) screen for evaluation of the functional effect of silencing each gene. Our selected endpoint was inhibition of growth. In rigorous postscreen validation processes, including quantitative RT-PCR, to ensure on-target silencing, deconvolution of pooled siRNAs and independent confirmation of effects with lentiviral short-hairpin RNA constructs, we identified a subset of genes required for mammary epithelial cell growth. Using three-dimensional Matrigel growth and differentiation assays and primary human mammary epithelial cell colony assays, we confirmed that these growth effects were not limited to the 184-hTERT cell line. We utilized the METABRIC dataset of 1,998 breast cancer patients to evaluate both the differential expression of these genes across breast cancer subtypes and their prognostic significance.ResultsWe identified 47 genes that are critically important for fibroblast-enhanced mammary epithelial cell growth. This group was enriched for several axonal guidance molecules and G protein–coupled receptors, as well as for the endothelin receptor PROCR. The majority of genes (43 of 47) identified in two dimensions were also required for three-dimensional growth, with HSD17B2, SNN and PROCR showing greater than tenfold reductions in acinar formation. Several genes, including PROCR and the neuronal pathfinding molecules EFNA4 and NTN1, were also required for proper differentiation and polarization in three-dimensional cultures. The 47 genes identified showed a significant nonrandom enrichment for differential expression among 10 molecular subtypes of breast cancer sampled from 1,998 patients. CD79A, SERPINH1, KCNJ5 and TMEM14C exhibited breast cancer subtype–independent overall survival differences.ConclusionDiverse transmembrane signals are required for mammary epithelial cell growth in two-dimensional and three-dimensional conditions. Strikingly, we define novel roles for axonal pathfinding receptors and ligands and the endothelin receptor in both growth and differentiation.

Collaboration


Dive into the Adrian Wan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew Roth

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Samuel Aparicio

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Ali Bashashati

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Karey Shumansky

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Allen W. Zhang

University of British Columbia

View shared research outputs
Researchain Logo
Decentralizing Knowledge