Network


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

Hotspot


Dive into the research topics where Petr Smirnov is active.

Publication


Featured researches published by Petr Smirnov.


Bioinformatics | 2016

PharmacoGx: an R package for analysis of large pharmacogenomic datasets

Petr Smirnov; Zhaleh Safikhani; Nehme El-Hachem; Dong Wang; Adrian She; Catharina Olsen; Mark Freeman; Heather Selby; Deena M.A. Gendoo; Patrick Grossmann; Andrew H. Beck; Hugo J.W.L. Aerts; Mathieu Lupien; Anna Goldenberg; Benjamin Haibe-Kains

UNLABELLED Pharmacogenomics holds great promise for the development of biomarkers of drug response and the design of new therapeutic options, which are key challenges in precision medicine. However, such data are scattered and lack standards for efficient access and analysis, consequently preventing the realization of the full potential of pharmacogenomics. To address these issues, we implemented PharmacoGx, an easy-to-use, open source package for integrative analysis of multiple pharmacogenomic datasets. We demonstrate the utility of our package in comparing large drug sensitivity datasets, such as the Genomics of Drug Sensitivity in Cancer and the Cancer Cell Line Encyclopedia. Moreover, we show how to use our package to easily perform Connectivity Map analysis. With increasing availability of drug-related data, our package will open new avenues of research for meta-analysis of pharmacogenomic data. AVAILABILITY AND IMPLEMENTATION PharmacoGx is implemented in R and can be easily installed on any system. The package is available from CRAN and its source code is available from GitHub. CONTACT [email protected] or [email protected] SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.


F1000Research | 2016

Assessment of pharmacogenomic agreement.

Zhaleh Safikhani; Nehme El-Hachem; Rene Quevedo; Petr Smirnov; Anna Goldenberg; Nicolai Juul Birkbak; Christopher E. Mason; Christos Hatzis; Leming Shi; Hugo J.W.L. Aerts; John Quackenbush; Benjamin Haibe-Kains

In 2013 we published an analysis demonstrating that drug response data and gene-drug associations reported in two independent large-scale pharmacogenomic screens, Genomics of Drug Sensitivity in Cancer (GDSC) and Cancer Cell Line Encyclopedia (CCLE), were inconsistent. The GDSC and CCLE investigators recently reported that their respective studies exhibit reasonable agreement and yield similar molecular predictors of drug response, seemingly contradicting our previous findings. Reanalyzing the authors’ published methods and results, we found that their analysis failed to account for variability in the genomic data and more importantly compared different drug sensitivity measures from each study, which substantially deviate from our more stringent consistency assessment. Our comparison of the most updated genomic and pharmacological data from the GDSC and CCLE confirms our published findings that the measures of drug response reported by these two groups are not consistent. We believe that a principled approach to assess the reproducibility of drug sensitivity predictors is necessary before envisioning their translation into clinical settings.


Cancer Research | 2017

Integrative Cancer Pharmacogenomics to Infer Large-Scale Drug Taxonomy

Nehme El-Hachem; Deena M.A. Gendoo; Laleh Soltan Ghoraie; Zhaleh Safikhani; Petr Smirnov; Christina Chung; Kenan Deng; Ailsa Fang; Erin Birkwood; Chantal Ho; Ruth Isserlin; Gary D. Bader; Anna Goldenberg; Benjamin Haibe-Kains

Identification of drug targets and mechanism of action (MoA) for new and uncharacterized anticancer drugs is important for optimization of treatment efficacy. Current MoA prediction largely relies on prior information including side effects, therapeutic indication, and chemoinformatics. Such information is not transferable or applicable for newly identified, previously uncharacterized small molecules. Therefore, a shift in the paradigm of MoA predictions is necessary toward development of unbiased approaches that can elucidate drug relationships and efficiently classify new compounds with basic input data. We propose here a new integrative computational pharmacogenomic approach, referred to as Drug Network Fusion (DNF), to infer scalable drug taxonomies that rely only on basic drug characteristics toward elucidating drug-drug relationships. DNF is the first framework to integrate drug structural information, high-throughput drug perturbation, and drug sensitivity profiles, enabling drug classification of new experimental compounds with minimal prior information. DNF taxonomy succeeded in identifying pertinent and novel drug-drug relationships, making it suitable for investigating experimental drugs with potential new targets or MoA. The scalability of DNF facilitated identification of key drug relationships across different drug categories, providing a flexible tool for potential clinical applications in precision medicine. Our results support DNF as a valuable resource to the cancer research community by providing new hypotheses on compound MoA and potential insights for drug repurposing. Cancer Res; 77(11); 3057-69. ©2017 AACR.


Nature Communications | 2017

Gene isoforms as expression-based biomarkers predictive of drug response in vitro

Zhaleh Safikhani; Petr Smirnov; Kelsie L. Thu; Jennifer Silvester; Nehme El-Hachem; Rene Quevedo; Mathieu Lupien; Tak W. Mak; David W. Cescon; Benjamin Haibe-Kains

Next-generation sequencing technologies have recently been used in pharmacogenomic studies to characterize large panels of cancer cell lines at the genomic and transcriptomic levels. Among these technologies, RNA-sequencing enable profiling of alternatively spliced transcripts. Given the high frequency of mRNA splicing in cancers, linking this feature to drug response will open new avenues of research in biomarker discovery. To identify robust transcriptomic biomarkers for drug response across studies, we develop a meta-analytical framework combining the pharmacological data from two large-scale drug screening datasets. We use an independent pan-cancer pharmacogenomic dataset to test the robustness of our candidate biomarkers across multiple cancer types. We further analyze two independent breast cancer datasets and find that specific isoforms of IGF2BP2, NECTIN4, ITGB6, and KLHDC9 are significantly associated with AZD6244, lapatinib, erlotinib, and paclitaxel, respectively. Our results support isoform expressions as a rich resource for biomarkers predictive of drug response.Altered mRNA splicing features in many cancers, but it has not been linked to drug response. Here, with their meta-analytic framework, the authors analyse pharmacogenomic data to identify isoform-based biomarkers predictive of in vitro drug response, and show them to frequently be strong predictors.


Proceedings of the National Academy of Sciences of the United States of America | 2018

Disruption of the anaphase-promoting complex confers resistance to TTK inhibitors in triple-negative breast cancer

Kelsie L. Thu; Jennifer Silvester; Meghan J. Elliott; W. Ba-alawi; M. H. Duncan; A. C. Elia; A. S. Mer; Petr Smirnov; Zhaleh Safikhani; Benjamin Haibe-Kains; Tak W. Mak; David W. Cescon

Significance Using functional genomic screens, we have identified resistance mechanisms to the clinical TTK protein kinase inhibitor (TTKi) CFI-402257 in breast cancer. As this and other TTKi are currently in clinical trials, understanding determinants of tumor drug response could permit rational selection of patients for treatment. We found that TTKi resistance is conferred by impairing anaphase-promoting complex/cyclosome (APC/C) function to minimize the lethal effects of mitotic segregation errors. Discovery of this mechanism in aneuploid cancer cells builds on previous reports indicating that weakening the APC/C promotes tolerance of chromosomal instability in diploid cells. Our work suggests that APC/C functional capacity may serve as a clinically useful biomarker of tumor response to TTKi that warrants investigation in ongoing clinical trials. TTK protein kinase (TTK), also known as Monopolar spindle 1 (MPS1), is a key regulator of the spindle assembly checkpoint (SAC), which functions to maintain genomic integrity. TTK has emerged as a promising therapeutic target in human cancers, including triple-negative breast cancer (TNBC). Several TTK inhibitors (TTKis) are being evaluated in clinical trials, and an understanding of the mechanisms mediating TTKi sensitivity and resistance could inform the successful development of this class of agents. We evaluated the cellular effects of the potent clinical TTKi CFI-402257 in TNBC models. CFI-402257 induced apoptosis and potentiated aneuploidy in TNBC lines by accelerating progression through mitosis and inducing mitotic segregation errors. We used genome-wide CRISPR/Cas9 screens in multiple TNBC cell lines to identify mechanisms of resistance to CFI-402257. Our functional genomic screens identified members of the anaphase-promoting complex/cyclosome (APC/C) complex, which promotes mitotic progression following inactivation of the SAC. Several screen candidates were validated to confer resistance to CFI-402257 and other TTKis using CRISPR/Cas9 and siRNA methods. These findings extend the observation that impairment of the APC/C enables cells to tolerate genomic instability caused by SAC inactivation, and support the notion that a measure of APC/C function could predict the response to TTK inhibition. Indeed, an APC/C gene expression signature is significantly associated with CFI-402257 response in breast and lung adenocarcinoma cell line panels. This expression signature, along with somatic alterations in genes involved in mitotic progression, represent potential biomarkers that could be evaluated in ongoing clinical trials of CFI-402257 or other TTKis.


Nucleic Acids Research | 2018

PharmacoDB: an integrative database for mining in vitro anticancer drug screening studies

Petr Smirnov; Victor Kofia; Alexander Maru; Mark Freeman; Chantal Ho; Nehme El-Hachem; George-Alexandru Adam; Wail Ba-alawi; Zhaleh Safikhani; Benjamin Haibe-Kains

Abstract Recent cancer pharmacogenomic studies profiled large panels of cell lines against hundreds of approved drugs and experimental chemical compounds. The overarching goal of these screens is to measure sensitivity of cell lines to chemical perturbations, correlate these measures to genomic features, and thereby develop novel predictors of drug response. However, leveraging these valuable data is challenging due to the lack of standards for annotating cell lines and chemical compounds, and quantifying drug response. Moreover, it has been recently shown that the complexity and complementarity of the experimental protocols used in the field result in high levels of technical and biological variation in the in vitro pharmacological profiles. There is therefore a need for new tools to facilitate rigorous comparison and integrative analysis of large-scale drug screening datasets. To address this issue, we have developed PharmacoDB (pharmacodb.pmgenomics.ca), a database integrating the largest cancer pharmacogenomic studies published to date. Here, we describe how the curation of cell line and chemical compound identifiers maximizes the overlap between datasets and how users can leverage such data to compare and extract robust drug phenotypes. PharmacoDB provides a unique resource to mine a compendium of curated cancer pharmacogenomic datasets that are otherwise disparate and difficult to integrate.


Physics in Medicine and Biology | 2017

Design of a HIFU array for the treatment of deep venous thrombosis: a simulation study

Petr Smirnov; Kullervo Hynynen

Deep venous thrombosis of the iliofemoral veins is a common and morbid disease, with the recommended interventional treatment carrying a high risk of hemorrhaging and complications. High intensity focused ultrasound delivered with a single element transducer has been shown to successfully precipitate thrombolysis non-invasively in vitro and in vivo. However, in all previous studies damage to the veins or surrounding tissue has been observed. Using a simulation model of the human thigh, this study investigated whether a phased array device could overcome the large focal region limitations faced by single transducer treatment devices. Effects of the size, shape and frequency of the array on its focal region were considered. It was found that a [Formula: see text] spaced array of 7680 elements operating at 500 kHz could consistently focus to a region fully contained within the femoral vein. Furthermore, it is possible to reduce the number of elements required by building arrays operating at lower frequencies. The results suggest that phased transducer arrays hold potential for developing a safe, non-invasive treatment of thrombolysis.


bioRxiv | 2018

Modeling cellular response in large-scale radiogenomic databases to advance precision radiotherapy

Venkata Sk Manem; Meghan Lambie; Petr Smirnov; Victor Kofia; Mark Freeman; Marianne Koritzinsky; M. Abazeed; Benjamin Haibe-Kains; Scott V. Bratman

Radiotherapy is integral to the care of a majority of cancer patients. Despite differences in tumor responses to radiation (radioresponse), dose prescriptions are not currently tailored to individual patients. Recent large-scale cancer cell line databases hold the promise of unravelling the complex molecular arrangements underlying cellular response to radiation, which is critical to novel predictive biomarker discovery. Here, we present RadioGx, a computational platform for integrative analyses of radioresponse using radiogenomic databases. We first used RadioGx to investigate the robustness of radioresponse assays and indicators. We then combined radioresponse and genome-wide molecular data with established radiobiological models to predict molecular pathways that are relevant for individual tissue types and conditions. We also applied RadioGx to pharmacogenomic data to identify several classes of drugs whose effects correlate with radioresponse. RadioGx provides a unique computational toolbox to advance preclinical research for radiation oncology and precision medicine.


Nature Communications | 2018

Author Correction: Gene isoforms as expression-based biomarkers predictive of drug response in vitro

Zhaleh Safikhani; Petr Smirnov; Kelsie L. Thu; Jennifer Silvester; Nehme El-Hachem; Rene Quevedo; Mathieu Lupien; Tak W. Mak; David W. Cescon; Benjamin Haibe-Kains

In the original version of this Article, financial support was not fully acknowledged. This error has now been corrected in both the PDF and HTML versions of the Article.


F1000Research | 2017

Revisiting inconsistency in large pharmacogenomic studies

Zhaleh Safikhani; Petr Smirnov; Mark Freeman; Nehme El-Hachem; Adrian She; Quevedo Rene; Anna Goldenberg; Nicolai Juul Birkbak; Christos Hatzis; Leming Shi; Andrew H. Beck; Hugo J.W.L. Aerts; John Quackenbush; Benjamin Haibe-Kains

Collaboration


Dive into the Petr Smirnov's collaboration.

Top Co-Authors

Avatar

Benjamin Haibe-Kains

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Zhaleh Safikhani

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mark Freeman

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Hugo J.W.L. Aerts

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David W. Cescon

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge