Network


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

Hotspot


Dive into the research topics where Arusha Oloumi is active.

Publication


Featured researches published by Arusha Oloumi.


Nature | 2012

The clonal and mutational evolution spectrum of primary triple-negative breast cancers.

Sohrab P. Shah; Andrew Roth; Rodrigo Goya; Arusha Oloumi; Gavin Ha; Yongjun Zhao; Gulisa Turashvili; Jiarui Ding; Kane Tse; Gholamreza Haffari; Ali Bashashati; Leah M Prentice; Jaswinder Khattra; Angela Burleigh; Damian Yap; Virginie Bernard; Andrew McPherson; Karey Shumansky; Anamaria Crisan; Ryan Giuliany; Alireza Heravi-Moussavi; Jamie Rosner; Daniel Lai; Inanc Birol; Richard Varhol; Angela Tam; Noreen Dhalla; Thomas Zeng; Kevin Ma; Simon K. Chan

Primary triple-negative breast cancers (TNBCs), a tumour type defined by lack of oestrogen receptor, progesterone receptor and ERBB2 gene amplification, represent approximately 16% of all breast cancers. Here we show in 104 TNBC cases that at the time of diagnosis these cancers exhibit a wide and continuous spectrum of genomic evolution, with some having only a handful of coding somatic aberrations in a few pathways, whereas others contain hundreds of coding somatic mutations. High-throughput RNA sequencing (RNA-seq) revealed that only approximately 36% of mutations are expressed. Using deep re-sequencing measurements of allelic abundance for 2,414 somatic mutations, we determine for the first time—to our knowledge—in an epithelial tumour subtype, the relative abundance of clonal frequencies among cases representative of the population. We show that TNBCs vary widely in their clonal frequencies at the time of diagnosis, with the basal subtype of TNBC showing more variation than non-basal TNBC. Although p53 (also known as TP53), PIK3CA and PTEN somatic mutations seem to be clonally dominant compared to other genes, in some tumours their clonal frequencies are incompatible with founder status. Mutations in cytoskeletal, cell shape and motility proteins occurred at lower clonal frequencies, suggesting that they occurred later during tumour progression. Taken together, our results show that understanding the biology and therapeutic responses of patients with TNBC will require the determination of individual tumour clonal genotypes.


The New England Journal of Medicine | 2010

ARID1A Mutations in Endometriosis-Associated Ovarian Carcinomas

Kimberly C. Wiegand; Sohrab P. Shah; Osama M. Al-Agha; Yongjun Zhao; Kane Tse; Thomas Zeng; Janine Senz; Melissa K. McConechy; Michael S. Anglesio; Steve E. Kalloger; Winnie Yang; Alireza Heravi-Moussavi; Ryan Giuliany; Christine Chow; John Fee; Abdalnasser Zayed; Leah M Prentice; Nataliya Melnyk; Gulisa Turashvili; Allen Delaney; Jason Madore; Stephen Yip; Andrew McPherson; Gavin Ha; Lynda Bell; Sian Fereday; Angela Tam; Laura Galletta; Patricia N. Tonin; Diane Provencher

BACKGROUND Ovarian clear-cell and endometrioid carcinomas may arise from endometriosis, but the molecular events involved in this transformation have not been described. METHODS We sequenced the whole transcriptomes of 18 ovarian clear-cell carcinomas and 1 ovarian clear-cell carcinoma cell line and found somatic mutations in ARID1A (the AT-rich interactive domain 1A [SWI-like] gene) in 6 of the samples. ARID1A encodes BAF250a, a key component of the SWI–SNF chromatin remodeling complex. We sequenced ARID1A in an additional 210 ovarian carcinomas and a second ovarian clear-cell carcinoma cell line and measured BAF250a expression by means of immunohistochemical analysis in an additional 455 ovarian carcinomas. RESULTS ARID1A mutations were seen in 55 of 119 ovarian clear-cell carcinomas (46%), 10 of 33 endometrioid carcinomas (30%), and none of the 76 high-grade serous ovarian carcinomas. Seventeen carcinomas had two somatic mutations each. Loss of the BAF250a protein correlated strongly with the ovarian clear-cell carcinoma and endometrioid carcinoma subtypes and the presence of ARID1A mutations. In two patients, ARID1A mutations and loss of BAF250a expression were evident in the tumor and contiguous atypical endometriosis but not in distant endometriotic lesions. CONCLUSIONS These data implicate ARID1A as a tumor-suppressor gene frequently disrupted in ovarian clear-cell and endometrioid carcinomas. Since ARID1A mutation and loss of BAF250a can be seen in the preneoplastic lesions, we speculate that this is an early event in the transformation of endometriosis into cancer. (Funded by the British Columbia Cancer Foundation and the Vancouver General Hospital–University of British Columbia Hospital Foundation.).


Cancer Research | 2011

Targeting tumor hypoxia: suppression of breast tumor growth and metastasis by novel carbonic anhydrase IX inhibitors.

Yuanmei Lou; Paul C. McDonald; Arusha Oloumi; Stephen Chia; Christina Ostlund; Ardalan Ahmadi; Alastair H. Kyle; Ulrich auf dem Keller; Samuel Leung; David Huntsman; Blaise Clarke; Brent W. Sutherland; Dawn Waterhouse; Marcel B. Bally; Calvin D. Roskelley; Christopher M. Overall; Andrew I. Minchinton; Fabio Pacchiano; Fabrizio Carta; Andrea Scozzafava; Nadia Touisni; Jean-Yves Winum; Claudiu T. Supuran; Shoukat Dedhar

Carbonic anhydrase IX (CAIX) is a hypoxia and HIF-1-inducible protein that regulates intra- and extracellular pH under hypoxic conditions and promotes tumor cell survival and invasion in hypoxic microenvironments. Interrogation of 3,630 human breast cancers provided definitive evidence of CAIX as an independent poor prognostic biomarker for distant metastases and survival. shRNA-mediated depletion of CAIX expression in 4T1 mouse metastatic breast cancer cells capable of inducing CAIX in hypoxia resulted in regression of orthotopic mammary tumors and inhibition of spontaneous lung metastasis formation. Stable depletion of CAIX in MDA-MB-231 human breast cancer xenografts also resulted in attenuation of primary tumor growth. CAIX depletion in the 4T1 cells led to caspase-independent cell death and reversal of extracellular acidosis under hypoxic conditions in vitro. Treatment of mice harboring CAIX-positive 4T1 mammary tumors with novel CAIX-specific small molecule inhibitors that mimicked the effects of CAIX depletion in vitro resulted in significant inhibition of tumor growth and metastasis formation in both spontaneous and experimental models of metastasis, without inhibitory effects on CAIX-negative tumors. Similar inhibitory effects on primary tumor growth were observed in mice harboring orthotopic tumors comprised of lung metatstatic MDA-MB-231 LM2-4(Luc+) cells. Our findings show that CAIX is vital for growth and metastasis of hypoxic breast tumors and is a specific, targetable biomarker for breast cancer metastasis.


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 Communications | 2016

The somatic mutation profiles of 2,433 breast cancers refines their genomic and transcriptomic landscapes

Bernard Pereira; Suet Feung Chin; Oscar M. Rueda; Hans Kristian Moen Vollan; Elena Provenzano; Helen Bardwell; Michelle Pugh; Linda Jones; Roslin Russell; Stephen John Sammut; Dana W.Y. Tsui; Bin Liu; Sarah-Jane Dawson; Jean Abraham; Helen Northen; John F. Peden; Abhik Mukherjee; Gulisa Turashvili; Andrew R. Green; Steve McKinney; Arusha Oloumi; Sohrab P. Shah; Nitzan Rosenfeld; Leigh C. Murphy; David R. Bentley; Ian O. Ellis; Arnie Purushotham; Sarah Pinder; Anne Lise Børresen-Dale; Helena M. Earl

The genomic landscape of breast cancer is complex, and inter- and intra-tumour heterogeneity are important challenges in treating the disease. In this study, we sequence 173 genes in 2,433 primary breast tumours that have copy number aberration (CNA), gene expression and long-term clinical follow-up data. We identify 40 mutation-driver (Mut-driver) genes, and determine associations between mutations, driver CNA profiles, clinical-pathological parameters and survival. We assess the clonal states of Mut-driver mutations, and estimate levels of intra-tumour heterogeneity using mutant-allele fractions. Associations between PIK3CA mutations and reduced survival are identified in three subgroups of ER-positive cancer (defined by amplification of 17q23, 11q13–14 or 8q24). High levels of intra-tumour heterogeneity are in general associated with a worse outcome, but highly aggressive tumours with 11q13–14 amplification have low levels of intra-tumour heterogeneity. These results emphasize the importance of genome-based stratification of breast cancer, and have important implications for designing therapeutic strategies.


Cancer Research | 2008

Rictor and Integrin-Linked Kinase Interact and Regulate Akt Phosphorylation and Cancer Cell Survival

Paul C. McDonald; Arusha Oloumi; Julia Mills; Iveta Dobreva; Mykola Maidan; Virginia Gray; Elizabeth D. Wederell; Marcel B. Bally; Leonard J. Foster; Shoukat Dedhar

An unbiased proteomic screen to identify integrin-linked kinase (ILK) interactors revealed rictor as an ILK-binding protein. This finding was interesting because rictor, originally identified as a regulator of cytoskeletal dynamics, is also a component of mammalian target of rapamycin complex 2 (mTORC2), a complex implicated in Akt phosphorylation. These functions overlap with known ILK functions. Coimmunoprecipitation analyses confirmed this interaction, and ILK and rictor colocalized in membrane ruffles and leading edges of cancer cells. Yeast two-hybrid assays showed a direct interaction between the NH(2)- and COOH-terminal domains of rictor and the ILK kinase domain. Depletion of ILK and rictor in breast and prostate cancer cell lines resulted in inhibition of Akt Ser(473) phosphorylation and induction of apoptosis, whereas, in several cell lines, depletion of mTOR increased Akt phosphorylation. Akt and Ser(473)P-Akt were detected in ILK immunoprecipitates and small interfering RNA-mediated depletion of rictor, but not mTOR, inhibited the amount of Ser(473)P-Akt in the ILK complex. Expression of the NH(2)-terminal (1-398 amino acids) rictor domain also resulted in the inhibition of ILK-associated Akt Ser(473) phosphorylation. These data show that rictor regulates the ability of ILK to promote Akt phosphorylation and cancer cell survival.


Nature Genetics | 2014

Ordering of mutations in preinvasive disease stages of esophageal carcinogenesis

Jamie M.J. Weaver; Caryn S. Ross-Innes; Nicholas Shannon; Andy G. Lynch; Tim Forshew; Mariagnese Barbera; Muhammed Murtaza; Chin-Ann J. Ong; Pierre Lao-Sirieix; Mark J. Dunning; Laura Smith; M.L.R. Smith; Charlotte Anderson; Benilton Carvalho; Maria O'Donovan; Timothy J. Underwood; Andrew May; Nicola Grehan; Richard H. Hardwick; Jim Davies; Arusha Oloumi; Sam Aparicio; Carlos Caldas; Matthew Eldridge; Paul A.W. Edwards; Nitzan Rosenfeld; Simon Tavaré; Rebecca C. Fitzgerald

Cancer genome sequencing studies have identified numerous driver genes, but the relative timing of mutations in carcinogenesis remains unclear. The gradual progression from premalignant Barretts esophagus to esophageal adenocarcinoma (EAC) provides an ideal model to study the ordering of somatic mutations. We identified recurrently mutated genes and assessed clonal structure using whole-genome sequencing and amplicon resequencing of 112 EACs. We next screened a cohort of 109 biopsies from 2 key transition points in the development of malignancy: benign metaplastic never-dysplastic Barretts esophagus (NDBE; n = 66) and high-grade dysplasia (HGD; n = 43). Unexpectedly, the majority of recurrently mutated genes in EAC were also mutated in NDBE. Only TP53 and SMAD4 mutations occurred in a stage-specific manner, confined to HGD and EAC, respectively. Finally, we applied this knowledge to identify high-risk Barretts esophagus in a new non-endoscopic test. In conclusion, mutations in EAC driver genes generally occur exceptionally early in disease development with profound implications for diagnostic and therapeutic strategies.


Bioinformatics | 2012

JointSNVMix: a probabilistic model for accurate detection of somatic mutations in normal/tumour paired next-generation sequencing data.

Andrew Roth; Jiarui Ding; Ryan D. Morin; Anamaria Crisan; Gavin Ha; Ryan Giuliany; Ali Bashashati; Martin Hirst; Gulisa Turashvili; Arusha Oloumi; Marco A. Marra; Samuel Aparicio; Sohrab P. Shah

Motivation: Identification of somatic single nucleotide variants (SNVs) in tumour genomes is a necessary step in defining the mutational landscapes of cancers. Experimental designs for genome-wide ascertainment of somatic mutations now routinely include next-generation sequencing (NGS) of tumour DNA and matched constitutional DNA from the same individual. This allows investigators to control for germline polymorphisms and distinguish somatic mutations that are unique to the tumour, thus reducing the burden of labour-intensive and expensive downstream experiments needed to verify initial predictions. In order to make full use of such paired datasets, computational tools for simultaneous analysis of tumour–normal paired sequence data are required, but are currently under-developed and under-represented in the bioinformatics literature. Results: In this contribution, we introduce two novel probabilistic graphical models called JointSNVMix1 and JointSNVMix2 for jointly analysing paired tumour–normal digital allelic count data from NGS experiments. In contrast to independent analysis of the tumour and normal data, our method allows statistical strength to be borrowed across the samples and therefore amplifies the statistical power to identify and distinguish both germline and somatic events in a unified probabilistic framework. Availability: The JointSNVMix models and four other models discussed in the article are part of the JointSNVMix software package available for download at http://compbio.bccrc.ca Contact: [email protected] Supplementary information:Supplementary data are available at Bioinformatics online.


Genome Research | 2012

Integrative analysis of genome-wide loss of heterozygosity and monoallelic expression at nucleotide resolution reveals disrupted pathways in triple-negative breast cancer

Gavin Ha; Andrew Roth; Daniel Lai; Ali Bashashati; Jiarui Ding; Rodrigo Goya; Ryan Giuliany; Jamie Rosner; Arusha Oloumi; Karey Shumansky; Suet-Feung Chin; Gulisa Turashvili; Martin Hirst; Carlos Caldas; Marco A. Marra; Samuel Aparicio; Sohrab P. Shah

Loss of heterozygosity (LOH) and copy number alteration (CNA) feature prominently in the somatic genomic landscape of tumors. As such, karyotypic aberrations in cancer genomes have been studied extensively to discover novel oncogenes and tumor-suppressor genes. Advances in sequencing technology have enabled the cost-effective detection of tumor genome and transcriptome mutation events at single-base-pair resolution; however, computational methods for predicting segmental regions of LOH in this context are not yet fully explored. Consequently, whole transcriptome, nucleotide-level resolution analysis of monoallelic expression patterns associated with LOH has not yet been undertaken in cancer. We developed a novel approach for inference of LOH from paired tumor/normal sequence data and applied it to a cohort of 23 triple-negative breast cancer (TNBC) genomes. Following extensive benchmarking experiments, we describe the nucleotide-resolution landscape of LOH in TNBC and assess the consequent effect of LOH on the transcriptomes of these tumors using RNA-seq-derived measurements of allele-specific expression. We show that the majority of monoallelic expression in the transcriptomes of triple-negative breast cancer can be explained by genomic regions of LOH and establish an upper bound for monoallelic expression that may be explained by other tumor-specific modifications such as epigenetics or mutations. Monoallelically expressed genes associated with LOH reveal that cell cycle, homologous recombination and actin-cytoskeletal functions are putatively disrupted by LOH in TNBC. Finally, we show how inference of LOH can be used to interpret allele frequencies of somatic mutations and postulate on temporal ordering of mutations in the evolutionary history of these tumors.


Oncogene | 2006

Modulation of Wnt3a-mediated nuclear |[beta]|-catenin accumulation and activation by integrin-linked kinase in mammalian cells

Arusha Oloumi; S Syam; Shoukat Dedhar

The Wnt gene family encodes secreted signaling molecules that play important roles in tumorgenesis and embryogenesis. The canonical Wnt signaling pathway regulates target gene expression via the stabilization and nuclear translocation of the cytoplasmic pool of β-catenin. The activation of integrin-linked kinase (ILK) is also known to regulate the stabilization and subsequent nuclear translocation of β-catenin in several epithelial cell models. We now report that molecular and pharmacological inhibition of ILK activity in mammalian cells directly modulates Wnt signaling by suppressing the stabilization and nuclear translocation of β-catenin, as well as β-catenin/Lef-mediated transcription. Inhibition of ILK activity, but not phosphatidylinositol-3 kinase (PI3K) or MEK activities suppresses nuclear β-catenin stabilization in cells stably expressing Wnt3a as well as in cells exposed to either Wnt3a conditioned media or purified Wnt3a. Furthermore, ILK inhibition reverses the Wnt3a-induced suppression of β-catenin phosphorylation that accompanies β-catenin stabilization. In addition, we show that ILK can be identified in a complex with Wnt pathway components such as adenomatous polyposis coli and GSK-3. Upon treatment of L cells with Wnt3a-CM, glycogen synthase kinase-3 (GSK-3β) becomes highly phosphorylated on Ser 9, which is completely abolished upon inhibition of ILK activity. However, acute exposure of L cells to purified Wnt3a does not result in the stimulation of GSK-3β Ser 9 phosphorylation, despite β-catenin stabilization. Together our data demonstrate that ILK activity can modulate acute Wnt3a mediated β-catenin phosphorylation, stabilization and nuclear activation in a PI3K-independent manner, as well as the more prolonged PI3K-dependent secondary effects of Wnt signaling on GSK-3 phosphorylation. Finally, we suggest that a novel small molecule inhibitor of ILK, QLT-0267, may be a useful tool in the regulation of pathological Wnt signaling.

Collaboration


Dive into the Arusha Oloumi's collaboration.

Top Co-Authors

Avatar

Shoukat Dedhar

Sunnybrook Health Sciences Centre

View shared research outputs
Top Co-Authors

Avatar

Sohrab P. Shah

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ali Bashashati

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Gulisa Turashvili

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Andrew Roth

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Jiarui Ding

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Marco A. Marra

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Martin Hirst

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Paul C. McDonald

University of British Columbia

View shared research outputs
Researchain Logo
Decentralizing Knowledge