scholarly journals Contextual Classifications of Cancer Driver Genes

2019 ◽  
Author(s):  
Pramod Chandrashekar ◽  
Navid Ahmadinejad ◽  
Junwen Wang ◽  
Aleksandar Sekulic ◽  
Jan B. Egan ◽  
...  

ABSTRACTFunctions of cancer driver genes depend on cellular contexts that vary substantially across tissues and organs. Distinguishing oncogenes (OGs) and tumor suppressor genes (TSGs) for each cancer type is critical to identifying clinically actionable targets. However, current resources for context-aware classifications of cancer drivers are limited. In this study, we show that the direction and magnitude of somatic selection of missense and truncating mutations of a gene are suggestive of its contextual activities. By integrating these features with ratiometric and conservation measures, we developed a computational method to categorize OGs and TSGs using exome sequencing data. This new method, named genes under selection in tumors (GUST) shows an overall accuracy of 0.94 when tested on manually curated benchmarks. Application of GUST to 10,172 tumor exomes of 33 cancer types identified 98 OGs and 179 TSGs, >70% of which promote tumorigenesis in only one cancer type. In broad-spectrum drivers shared across multiple cancer types, we found heterogeneous mutational hotspots modifying distinct functional domains, implicating the synchrony of convergent and divergent disease mechanisms. We further discovered two novel OGs and 28 novel TSGs with high confidence. The GUST program is available at https://github.com/liliulab/gust. A database with pre-computed classifications is available at https://liliulab.shinyapps.io/gust

2022 ◽  
Author(s):  
Malvika Sudhakar ◽  
Raghunathan Rengaswamy ◽  
Karthik Raman

The progression of tumorigenesis starts with a few mutational and structural driver events in the cell. Various cohort-based computational tools exist to identify driver genes but require a large number of samples to produce reliable results. Many studies use different methods to identify driver mutations/genes from mutations that have no impact on tumour progression; however, a small fraction of patients show no mutational events in any known driver genes. Current unsupervised methods map somatic and expression data onto a network to identify the perturbation in the network. Our method is the first machine learning model to classify genes as tumour suppressor gene (TSG), oncogene (OG) or neutral, thus assigning the functional impact of the gene in the patient. In this study, we develop a multi-omic approach, PIVOT (Personalised Identification of driVer OGs and TSGs), to train on experimentally or computationally validated mutational and structural driver events. Given the lack of any gold standards for the identification of personalised driver genes, we label the data using four strategies and, based on classification metrics, show gene-based labelling strategies perform best. We build different models using SNV, RNA, and multi-omic features to be used based on the data available. Our models trained on multi-omic data improved predictions compared to mutation and expression data, achieving an accuracy >0.99 for BRCA, LUAD and COAD datasets. We show network and expression-based features contribute the most to PIVOT. Our predictions on BRCA, COAD and LUAD cancer types reveal commonly altered genes such as TP53, and PIK3CA, which are predicted drivers for multiple cancer types. Along with known driver genes, our models also identify new driver genes such as PRKCA, SOX9 and PSMD4. Our multi-omic model labels both CNV and mutations with a more considerable contribution by CNV alterations. While predicting labels for genes mutated in multiple samples, we also label rare driver events occurring in as few as one sample. We also identify genes with dual roles within the same cancer type. Overall, PIVOT labels personalised driver genes as TSGs and OGs and also identifies rare driver genes. PIVOT is available at https://github.com/RamanLab/PIVOT.


2020 ◽  
Vol 3 (1) ◽  
Author(s):  
Xiaobao Dong ◽  
Dandan Huang ◽  
Xianfu Yi ◽  
Shijie Zhang ◽  
Zhao Wang ◽  
...  

AbstractMutation-specific effects of cancer driver genes influence drug responses and the success of clinical trials. We reasoned that these effects could unbalance the distribution of each mutation across different cancer types, as a result, the cancer preference can be used to distinguish the effects of the causal mutation. Here, we developed a network-based framework to systematically measure cancer diversity for each driver mutation. We found that half of the driver genes harbor cancer type-specific and pancancer mutations simultaneously, suggesting that the pervasive functional heterogeneity of the mutations from even the same driver gene. We further demonstrated that the specificity of the mutations could influence patient drug responses. Moreover, we observed that diversity was generally increased in advanced tumors. Finally, we scanned potentially novel cancer driver genes based on the diversity spectrum. Diversity spectrum analysis provides a new approach to define driver mutations and optimize off-label clinical trials.


2017 ◽  
Author(s):  
Luis Zapata ◽  
Hana Susak ◽  
Oliver Drechsel ◽  
Marc R. Friedländer ◽  
Xavier Estivill ◽  
...  

AbstractTumors are composed of an evolving population of cells subjected to tissue-specific selection, which fuels tumor heterogeneity and ultimately complicates cancer driver gene identification. Here, we integrate cancer cell fraction, population recurrence, and functional impact of somatic mutations as signatures of selection into a Bayesian inference model for driver prediction. In an in-depth benchmark, we demonstrate that our model, cDriver, outperforms competing methods when analyzing solid tumors, hematological malignancies, and pan-cancer datasets. Applying cDriver to exome sequencing data of 21 cancer types from 6,870 individuals revealed 98 unreported tumor type-driver gene connections. These novel connections are highly enriched for chromatin-modifying proteins, hinting at a universal role of chromatin regulation in cancer etiology. Although infrequently mutated as single genes, we show that chromatin modifiers are altered in a large fraction of cancer patients. In summary, we demonstrate that integration of evolutionary signatures is key for identifying mutational driver genes, thereby facilitating the discovery of novel therapeutic targets for cancer treatment.


2019 ◽  
Author(s):  
Pramod Chandrashekar ◽  
Navid Ahmadinejad ◽  
Junwen Wang ◽  
Aleksandar Sekulic ◽  
Jan B Egan ◽  
...  

Abstract Motivation Functions of cancer driver genes vary substantially across tissues and organs. Distinguishing passenger genes (PGs), oncogenes (OGs) and tumor suppressor genes (TSGs) for each cancer type is critical for understanding tumor biology and identifying clinically actionable targets. Although many computational tools are available to predict putative cancer driver genes, resources for context-aware classifications of OGs and TSGs are limited. Results We show that the direction and magnitude of somatic selection of protein-coding mutations are significantly different for PGs, OGs and TSGs. Based on these patterns, we develop a new method (genes under selection in tumors, GUST) to discover OGs and TSGs in a cancer-type specific manner. GUST shows a high accuracy (92%) when evaluated via strict cross-validations. Its application to 10,172 tumor exomes found known and novel cancer drivers with high tissue-specificities. In 11 out of 13 OGs shared among multiple cancer types, we found functional domains selectively engaged in different cancers, suggesting differences in disease mechanisms. Availability An R implementation of the GUST algorithm is available at https://github.com/liliulab/gust. A database with pre-computed results is available at https://liliulab.shinyapps.io/gust. Supplementary information Supplementary data are available at Bioinformatics online.


2016 ◽  
Vol 6 (1) ◽  
Author(s):  
Ho Jang ◽  
Youngmi Hur ◽  
Hyunju Lee

Abstract DNA copy number alterations (CNAs) are the main genomic events that occur during the initiation and development of cancer. Distinguishing driver aberrant regions from passenger regions, which might contain candidate target genes for cancer therapies, is an important issue. Several methods for identifying cancer-driver genes from multiple cancer patients have been developed for single nucleotide polymorphism (SNP) arrays. However, for NGS data, methods for the SNP array cannot be directly applied because of different characteristics of NGS such as higher resolutions of data without predefined probes and incorrectly mapped reads to reference genomes. In this study, we developed a wavelet-based method for identification of focal genomic alterations for sequencing data (WIFA-Seq). We applied WIFA-Seq to whole genome sequencing data from glioblastoma multiforme, ovarian serous cystadenocarcinoma and lung adenocarcinoma, and identified focal genomic alterations, which contain candidate cancer-related genes as well as previously known cancer-driver genes.


2016 ◽  
Vol 14 (06) ◽  
pp. 1650031 ◽  
Author(s):  
Ana B. Pavel ◽  
Cristian I. Vasile

Cancer is a complex and heterogeneous genetic disease. Different mutations and dysregulated molecular mechanisms alter the pathways that lead to cell proliferation. In this paper, we explore a method which classifies genes into oncogenes (ONGs) and tumor suppressors. We optimize this method to identify specific (ONGs) and tumor suppressors for breast cancer, lung adenocarcinoma (LUAD), lung squamous cell carcinoma (LUSC) and colon adenocarcinoma (COAD), using data from the cancer genome atlas (TCGA). A set of genes were previously classified as ONGs and tumor suppressors across multiple cancer types (Science 2013). Each gene was assigned an ONG score and a tumor suppressor score based on the frequency of its driver mutations across all variants from the catalogue of somatic mutations in cancer (COSMIC). We evaluate and optimize this approach within different cancer types from TCGA. We are able to determine known driver genes for each of the four cancer types. After establishing the baseline parameters for each cancer type, we identify new driver genes for each cancer type, and the molecular pathways that are highly affected by them. Our methodology is general and can be applied to different cancer subtypes to identify specific driver genes and improve personalized therapy.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Sharaf J. Malebary ◽  
Yaser Daanial Khan

AbstractCancer is driven by distinctive sorts of changes and basic variations in genes. Recognizing cancer driver genes is basic for accurate oncological analysis. Numerous methodologies to distinguish and identify drivers presently exist, but efficient tools to combine and optimize them on huge datasets are few. Most strategies for prioritizing transformations depend basically on frequency-based criteria. Strategies are required to dependably prioritize organically dynamic driver changes over inert passengers in high-throughput sequencing cancer information sets. This study proposes a model namely PCDG-Pred which works as a utility capable of distinguishing cancer driver and passenger attributes of genes based on sequencing data. Keeping in view the significance of the cancer driver genes an efficient method is proposed to identify the cancer driver genes. Further, various validation techniques are applied at different levels to establish the effectiveness of the model and to obtain metrics like accuracy, Mathew’s correlation coefficient, sensitivity, and specificity. The results of the study strongly indicate that the proposed strategy provides a fundamental functional advantage over other existing strategies for cancer driver genes identification. Subsequently, careful experiments exhibit that the accuracy metrics obtained for self-consistency, independent set, and cross-validation tests are 91.08%., 87.26%, and 92.48% respectively.


2021 ◽  
Author(s):  
Chenye Wang ◽  
Junhan Shi ◽  
Jiansheng Cai ◽  
Yusen Zhang ◽  
Xiaoqi Zheng ◽  
...  

Abstract Background: Recent advances in next-generation sequencing technologies have helped investigators generate massive amounts of cancer genomic data. A critical challenge in cancer genomics is identification of a few driver mutation genes from a much larger number of passenger mutation genes. However, majority of existing computational approaches underuse the co-occurrence information of the individuals, which deems to be important in tumorigenesis and tumor progression. Driver gene list predicted from these tools are prone to be false positive, recent research is far from achieving the ultimate goal of discovering a complete catalog of driver genes. Results: To make full use of co-mutation information, we present a random walk algorithm referred to as DriverRWH on a weighted gene mutation hypergraph model, using somatic mutation data and molecular interaction network data to prioritize candidate driver genes. Applied to tumor samples of different cancer types from The Cancer Genome Atlas (TCGA), DriverRWH shows significantly better performance than state-of-art prioritization methods in terms of the area under the curve (AUC) scores and the cumulative number of known driver genes recovered in top-ranked candidate genes. DriverRWH recovers approximately 50% known driver genes in the top 30 ranked candidate genes for more than half of the cancer types. In addition, DriverRWH is also highly robust to perturbations in the mutation data and gene functional network data. Conclusion: DriverRWH is effective among various cancer types in prioritizes cancer driver genes and provides considerable improvement over other tools with a better balance of precision and sensitivity. It can be a useful tool for detecting potential driver genes and facilitate targeted cancer therapies.


Sign in / Sign up

Export Citation Format

Share Document