Network


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

Hotspot


Dive into the research topics where Selcuk Onur Sumer is active.

Publication


Featured researches published by Selcuk Onur Sumer.


Cancer Discovery | 2012

The cBio Cancer Genomics Portal: An Open Platform for Exploring Multidimensional Cancer Genomics Data

Ethan Cerami; Jianjiong Gao; Ugur Dogrusoz; Benjamin E. Gross; Selcuk Onur Sumer; Bülent Arman Aksoy; Anders Jacobsen; Caitlin J. Byrne; Michael L. Heuer; Erik G. Larsson; Yevgeniy Antipin; Boris Reva; Arthur P. Goldberg; Chris Sander; Nikolaus Schultz

The cBio Cancer Genomics Portal (http://cbioportal.org) is an open-access resource for interactive exploration of multidimensional cancer genomics data sets, currently providing access to data from more than 5,000 tumor samples from 20 cancer studies. The cBio Cancer Genomics Portal significantly lowers the barriers between complex genomic data and cancer researchers who want rapid, intuitive, and high-quality access to molecular profiles and clinical attributes from large-scale cancer genomics projects and empowers researchers to translate these rich data sets into biologic insights and clinical applications.


Science Signaling | 2013

Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal.

Jian Jiong Gao; Bülent Arman Aksoy; Ugur Dogrusoz; Gideon Dresdner; Benjamin E. Gross; Selcuk Onur Sumer; Yichao Sun; Anders Jacobsen; Rileen Sinha; Erik Larsson; Ethan Cerami; Chris Sander; Nikolaus Schultz

The cBioPortal enables integration, visualization, and analysis of multidimensional cancer genomic and clinical data. The cBioPortal for Cancer Genomics (http://cbioportal.org) provides a Web resource for exploring, visualizing, and analyzing multidimensional cancer genomics data. The portal reduces molecular profiling data from cancer tissues and cell lines into readily understandable genetic, epigenetic, gene expression, and proteomic events. The query interface combined with customized data storage enables researchers to interactively explore genetic alterations across samples, genes, and pathways and, when available in the underlying data, to link these to clinical outcomes. The portal provides graphical summaries of gene-level data from multiple platforms, network visualization and analysis, survival analysis, patient-centric queries, and software programmatic access. The intuitive Web interface of the portal makes complex cancer genomics profiles accessible to researchers and clinicians without requiring bioinformatics expertise, thus facilitating biological discoveries. Here, we provide a practical guide to the analysis and visualization features of the cBioPortal for Cancer Genomics.


Genome Medicine | 2017

3D clusters of somatic mutations in cancer reveal numerous rare mutations as functional targets

Jianjiong Gao; Matthew T. Chang; Hannah Johnsen; Sizhi Paul Gao; Brooke E. Sylvester; Selcuk Onur Sumer; Hongxin Zhang; David B. Solit; Barry S. Taylor; Nikolaus Schultz; Chris Sander

Many mutations in cancer are of unknown functional significance. Standard methods use statistically significant recurrence of mutations in tumor samples as an indicator of functional impact. We extend such analyses into the long tail of rare mutations by considering recurrence of mutations in clusters of spatially close residues in protein structures. Analyzing 10,000 tumor exomes, we identify more than 3000 rarely mutated residues in proteins as potentially functional and experimentally validate several in RAC1 and MAP2K1. These potential driver mutations (web resources: 3dhotspots.org and cBioPortal.org) can extend the scope of genomically informed clinical trials and of personalized choice of therapy.


eLife | 2015

Perturbation biology nominates upstream–downstream drug combinations in RAF inhibitor resistant melanoma cells

Anil Korkut; Weiqing Wang; Emek Demir; Bülent Arman Aksoy; Xiaohong Jing; Evan Molinelli; Özgün Babur; Debra Bemis; Selcuk Onur Sumer; David B. Solit; Christine A. Pratilas; Chris Sander

Resistance to targeted cancer therapies is an important clinical problem. The discovery of anti-resistance drug combinations is challenging as resistance can arise by diverse escape mechanisms. To address this challenge, we improved and applied the experimental-computational perturbation biology method. Using statistical inference, we build network models from high-throughput measurements of molecular and phenotypic responses to combinatorial targeted perturbations. The models are computationally executed to predict the effects of thousands of untested perturbations. In RAF-inhibitor resistant melanoma cells, we measured 143 proteomic/phenotypic entities under 89 perturbation conditions and predicted c-Myc as an effective therapeutic co-target with BRAF or MEK. Experiments using the BET bromodomain inhibitor JQ1 affecting the level of c-Myc protein and protein kinase inhibitors targeting the ERK pathway confirmed the prediction. In conclusion, we propose an anti-cancer strategy of co-targeting a specific upstream alteration and a general downstream point of vulnerability to prevent or overcome resistance to targeted drugs. DOI: http://dx.doi.org/10.7554/eLife.04640.001


PLOS Computational Biology | 2016

A Multi-Method Approach for Proteomic Network Inference in 11 Human Cancers.

Yasin Şenbabaoğlu; Selcuk Onur Sumer; Francisco Sanchez-Vega; Debra Bemis; Giovanni Ciriello; Nikolaus Schultz; Chris Sander

Protein expression and post-translational modification levels are tightly regulated in neoplastic cells to maintain cellular processes known as ‘cancer hallmarks’. The first Pan-Cancer initiative of The Cancer Genome Atlas (TCGA) Research Network has aggregated protein expression profiles for 3,467 patient samples from 11 tumor types using the antibody based reverse phase protein array (RPPA) technology. The resultant proteomic data can be utilized to computationally infer protein-protein interaction (PPI) networks and to study the commonalities and differences across tumor types. In this study, we compare the performance of 13 established network inference methods in their capacity to retrieve the curated Pathway Commons interactions from RPPA data. We observe that no single method has the best performance in all tumor types, but a group of six methods, including diverse techniques such as correlation, mutual information, and regression, consistently rank highly among the tested methods. We utilize the high performing methods to obtain a consensus network; and identify four robust and densely connected modules that reveal biological processes as well as suggest antibody–related technical biases. Mapping the consensus network interactions to Reactome gene lists confirms the pan-cancer importance of signal transduction pathways, innate and adaptive immune signaling, cell cycle, metabolism, and DNA repair; and also suggests several biological processes that may be specific to a subset of tumor types. Our results illustrate the utility of the RPPA platform as a tool to study proteomic networks in cancer.


Bioinformatics | 2014

Pattern search in BioPAX models

Özgün Babur; Bülent Arman Aksoy; Igor Rodchenkov; Selcuk Onur Sumer; Chris Sander; Emek Demir

Motivation: BioPAX is a standard language for representing complex cellular processes, including metabolic networks, signal transduction and gene regulation. Owing to the inherent complexity of a BioPAX model, searching for a specific type of subnetwork can be non-trivial and difficult. Results: We developed an open source and extensible framework for defining and searching graph patterns in BioPAX models. We demonstrate its use with a sample pattern that captures directed signaling relations between proteins. We provide search results for the pattern obtained from the Pathway Commons database and compare these results with the current data in signaling databases SPIKE and SignaLink. Results show that a pattern search in public pathway data can identify a substantial amount of signaling relations that do not exist in signaling databases. Availability: BioPAX-pattern software was developed in Java. Source code and documentation is freely available at http://code.google.com/p/biopax-pattern under Lesser GNU Public License. Contact: [email protected] Supplementary information: Supplementary data are available at Bioinformatics online.


PLOS ONE | 2015

SBGNViz: A Tool for Visualization and Complexity Management of SBGN Process Description Maps.

Mecit Sari; Istemi Bahceci; Ugur Dogrusoz; Selcuk Onur Sumer; Bülent Arman Aksoy; Özgün Babur; Emek Demir

Background Information about cellular processes and pathways is becoming increasingly available in detailed, computable standard formats such as BioPAX and SBGN. Effective visualization of this information is a key recurring requirement for biological data analysis, especially for -omic data. Biological data analysis is rapidly migrating to web based platforms; thus there is a substantial need for sophisticated web based pathway viewers that support these platforms and other use cases. Results Towards this goal, we developed a web based viewer named SBGNViz for process description maps in SBGN (SBGN-PD). SBGNViz can visualize both BioPAX and SBGN formats. Unique features of SBGNViz include the ability to nest nodes to arbitrary depths to represent molecular complexes and cellular locations, automatic pathway layout, editing and highlighting facilities to enable focus on sub-maps, and the ability to inspect pathway members for detailed information from EntrezGene. SBGNViz can be used within a web browser without any installation and can be readily embedded into web pages. SBGNViz has two editions built with ActionScript and JavaScript. The JavaScript edition, which also works on touch enabled devices, introduces novel methods for managing and reducing complexity of large SBGN-PD maps for more effective analysis. Conclusion SBGNViz fills an important gap by making the large and fast-growing corpus of rich pathway information accessible to web based platforms. SBGNViz can be used in a variety of contexts and in multiple scenarios ranging from visualization of the results of a single study in a web page to building data analysis platforms.


Cancer Discovery | 2017

Accelerating Discovery of Functional Mutant Alleles in Cancer

Matthew T. Chang; Tripti Shrestha Bhattarai; Alison M. Schram; Craig M. Bielski; Mark T.A. Donoghue; Philip Jonsson; Debyani Chakravarty; Sarah Phillips; Cyriac Kandoth; Alexander Penson; Alexander N. Gorelick; Tambudzai Shamu; Swati Patel; Christopher C. Harris; Jianjiong Gao; Selcuk Onur Sumer; Ritika Kundra; Pedram Razavi; Bob T. Li; Dalicia Reales; Nicholas D. Socci; Gowtham Jayakumaran; Ahmet Zehir; Ryma Benayed; Maria E. Arcila; Sarat Chandarlapaty; Marc Ladanyi; Nikolaus Schultz; José Baselga; Michael F. Berger

Most mutations in cancer are rare, which complicates the identification of therapeutically significant mutations and thus limits the clinical impact of genomic profiling in patients with cancer. Here, we analyzed 24,592 cancers including 10,336 prospectively sequenced patients with advanced disease to identify mutant residues arising more frequently than expected in the absence of selection. We identified 1,165 statistically significant hotspot mutations of which 80% arose in 1 in 1,000 or fewer patients. Of 55 recurrent in-frame indels, we validated that novel AKT1 duplications induced pathway hyperactivation and conferred AKT inhibitor sensitivity. Cancer genes exhibit different rates of hotspot discovery with increasing sample size, with few approaching saturation. Consequently, 26% of all hotspots in therapeutically actionable oncogenes were novel. Upon matching a subset of affected patients directly to molecularly targeted therapy, we observed radiographic and clinical responses. Population-scale mutant allele discovery illustrates how the identification of driver mutations in cancer is far from complete.Significance: Our systematic computational, experimental, and clinical analysis of hotspot mutations in approximately 25,000 human cancers demonstrates that the long right tail of biologically and therapeutically significant mutant alleles is still incompletely characterized. Sharing prospective genomic data will accelerate hotspot identification, thereby expanding the reach of precision oncology in patients with cancer. Cancer Discov; 8(2); 174-83. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 127.


Nucleic Acids Research | 2016

MutationAligner: a resource of recurrent mutation hotspots in protein domains in cancer.

Nicholas Paul Gauthier; Ed Reznik; Jianjiong Gao; Selcuk Onur Sumer; Nikolaus Schultz; Chris Sander; Martin L. Miller

The MutationAligner web resource, available at http://www.mutationaligner.org, enables discovery and exploration of somatic mutation hotspots identified in protein domains in currently (mid-2015) more than 5000 cancer patient samples across 22 different tumor types. Using multiple sequence alignments of protein domains in the human genome, we extend the principle of recurrence analysis by aggregating mutations in homologous positions across sets of paralogous genes. Protein domain analysis enhances the statistical power to detect cancer-relevant mutations and links mutations to the specific biological functions encoded in domains. We illustrate how the MutationAligner database and interactive web tool can be used to explore, visualize and analyze mutation hotspots in protein domains across genes and tumor types. We believe that MutationAligner will be an important resource for the cancer research community by providing detailed clues for the functional importance of particular mutations, as well as for the design of functional genomics experiments and for decision support in precision medicine. MutationAligner is slated to be periodically updated to incorporate additional analyses and new data from cancer genomics projects.


Cancer Research | 2013

Abstract 5140: Individual patient cancer profiles in the cBio Cancer Genomic Portal.

Jianjiong Gao; Selcuk Onur Sumer; Gideon Dresdner; Bülent Arman Aksoy; Chris Sander; Nikolaus Schultz

The most prominent or most interesting genomic alteration events from an individual tumor sample can now be browsed and analyzed in the cBio Cancer Genomics Portal. With the rapid accumulation of detailed and comprehensive genomic maps of thousands of tumors in The Cancer Genome Atlas and other projects it has now become feasible to nominate the functionally most significant events affecting a tumor from an individual patient. The cBio Cancer Genomics Portal (http://cbioportal.org) allows interactive exploration of multidimensional cancer genomics data sets, currently for more than 6,000 tumor samples from 20 cancer genomics studies, including all TCGA projects. This information gateway significantly lowers the barrier between complex genomic data and their efficient use by cancer researchers for the development of biologic insights and clinical applications. In addition to gene-by-gene alteration maps across many samples and across diverse tumor types, one can now view genomic alterations in individual tumor samples. As there are potentially hundreds or thousands of genomic alterations in any single tumor sample, it is crucially important to select, for inspection and analysis, alteration events most likely to contribute to oncogenesis or affect the response to therapy. In the cBio portal patient view, this selection is done making use of recurrence statistics, background functional knowledge and predicted functional impact, under the control of the cancer researcher. All relevant data about a tumor are displayed on a single page, including clinical characteristics, summaries of the extent of mutations and copy-number alterations, as well as details about mutated, amplified, and deleted genes. Genomic alterations are filtered by the following criteria: recurrence of mutations or copy-number alterations across the tumor cohort (MutSig and GISTIC), mutation occurrence in COSMIC, and by cancer gene annotation (via the Sanger Cancer Gene Census and other sources). The patient view also provides information about drugs that target the altered genes and lists relevant clinical trials. The patient view is fully interactive and enables quick and easy assessment of all relevant genomic events in individual tumor samples. All data can be viewed in the context of the other tumors in the cohort, which facilitates classification of tumor samples by genomic criteria and can supplement standard pathology. Once characterization of genomic alterations in tumor samples becomes standard practice in patient care, this tool could be used to assess prognosis and guide treatment decisions, ideally the personalized choice of targeted therapies. Citation Format: Jianjiong Gao, Selcuk Onur Sumer, Gideon Dresdner, Bulent Arman Aksoy, Chris Sander, Nikolaus Schultz. Individual patient cancer profiles in the cBio Cancer Genomic Portal. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 5140. doi:10.1158/1538-7445.AM2013-5140

Collaboration


Dive into the Selcuk Onur Sumer's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nikolaus Schultz

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jianjiong Gao

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Bülent Arman Aksoy

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Benjamin E. Gross

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emek Demir

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hongxin Zhang

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Matthew T. Chang

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Özgün Babur

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge