drug sensitivity prediction
Recently Published Documents


TOTAL DOCUMENTS

62
(FIVE YEARS 31)

H-INDEX

12
(FIVE YEARS 4)

2022 ◽  
Vol 11 ◽  
Author(s):  
Xiya Jia ◽  
Bing Chen ◽  
Ziteng Li ◽  
Shenglin Huang ◽  
Siyuan Chen ◽  
...  

BackgroundGastric cancer (GC) is a highly molecular heterogeneous tumor with poor prognosis. Epithelial-mesenchymal transition (EMT) process and cancer stem cells (CSCs) are reported to share common signaling pathways and cause poor prognosis in GC. Considering about the close relationship between these two processes, we aimed to establish a gene signature based on both processes to achieve better prognostic prediction in GC.MethodsThe gene signature was constructed by univariate Cox and the least absolute shrinkage and selection operator (LASSO) Cox regression analyses by using The Cancer Genome Atlas (TCGA) GC cohort. We performed enrichment analyses to explore the potential mechanisms of the gene signature. Kaplan-Meier analysis and time-dependent receiver operating characteristic (ROC) curves were implemented to assess its prognostic value in TCGA cohort. The prognostic value of gene signature on overall survival (OS), disease-free survival (DFS), and drug sensitivity was validated in different cohorts. Quantitative reverse transcription polymerase chain reaction (RT-qPCR) validation of the prognostic value of gene signature for OS and DFS prediction was performed in the Fudan cohort.ResultsA prognostic signature including SERPINE1, EDIL3, RGS4, and MATN3 (SERM signature) was constructed to predict OS, DFS, and drug sensitivity in GC. Enrichment analyses illustrated that the gene signature has tight connection with the CSC and EMT processes in GC. Patients were divided into two groups based on the risk score obtained from the formula. The Kaplan-Meier analyses indicated high-risk group yielded significantly poor prognosis compared with low-risk group. Pearson’s correlation analysis indicated that the risk score was positively correlated with carboplatin and 5-fluorouracil IC50 of GC cell lines. Multivariate Cox regression analyses showed that the gene signature was an independent prognostic factor for predicting GC patients’ OS, DFS, and susceptibility to adjuvant chemotherapy.ConclusionsOur SERM prognostic signature is of great value for OS, DFS, and drug sensitivity prediction in GC, which may give guidance to the development of targeted therapy for CSC- and EMT-related gene in the future.


2021 ◽  
Vol 12 ◽  
Author(s):  
Yaojia Chen ◽  
Liran Juan ◽  
Xiao Lv ◽  
Lei Shi

Modeling-based anti-cancer drug sensitivity prediction has been extensively studied in recent years. While most drug sensitivity prediction models only use gene expression data, the remarkable impacts of gene mutation, methylation, and copy number variation on drug sensitivity are neglected. Drug sensitivity prediction can both help protect patients from some adverse drug reactions and improve the efficacy of treatment. Genomics data are extremely useful for drug sensitivity prediction task. This article reviews the role of drug sensitivity prediction, describes a variety of methods for predicting drug sensitivity. Moreover, the research significance of drug sensitivity prediction, as well as existing problems are well discussed.


2021 ◽  
pp. 145-154
Author(s):  
Delora Baptista ◽  
João Correia ◽  
Bruno Pereira ◽  
Miguel Rocha

2021 ◽  
Author(s):  
Hossein Sharifi-Noghabi ◽  
Soheil Jahangiri-Tazehkand ◽  
Casey Hon ◽  
Petr Smirnov ◽  
Anthony Mammoliti ◽  
...  

ABSTRACTThe goal of precision oncology is to tailor treatment for patients individually using the genomic profile of their tumors. Pharmacogenomics datasets such as cancer cell lines are among the most valuable resources for drug sensitivity prediction, a crucial task of precision oncology. Machine learning methods have been employed to predict drug sensitivity based on the multiple omics data available for large panels of cancer cell lines. However, there are no comprehensive guidelines on how to properly train and validate such machine learning models for drug sensitivity prediction. In this paper, we introduce a set of guidelines for different aspects of training a predictor using cell line datasets. These guidelines provide extensive analysis of the generalization of drug sensitivity predictors, and challenge many current practices in the community including the choice of training dataset and measure of drug sensitivity. Application of these guidelines in future studies will enable the development of more robust preclinical biomarkers.


Cell Systems ◽  
2021 ◽  
Author(s):  
Marco Tognetti ◽  
Attila Gabor ◽  
Mi Yang ◽  
Valentina Cappelletti ◽  
Jonas Windhager ◽  
...  

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Yi-Ching Tang ◽  
Assaf Gottlieb

AbstractComputational approaches to predict drug sensitivity can promote precision anticancer therapeutics. Generalizable and explainable models are of critical importance for translation to guide personalized treatment and are often overlooked in favor of prediction performance. Here, we propose PathDSP: a pathway-based model for drug sensitivity prediction that integrates chemical structure information with enrichment of cancer signaling pathways across drug-associated genes, gene expression, mutation and copy number variation data to predict drug response on the Genomics of Drug Sensitivity in Cancer dataset. Using a deep neural network, we outperform state-of-the-art deep learning models, while demonstrating good generalizability a separate dataset of the Cancer Cell Line Encyclopedia as well as provide explainable results, demonstrated through case studies that are in line with current knowledge. Additionally, our pathway-based model achieved a good performance when predicting unseen drugs and cells, with potential utility for drug development and for guiding individualized medicine.


2021 ◽  
Author(s):  
Krzysztof Koras ◽  
Ewa Kizling ◽  
Dilafruz Juraeva ◽  
Eike Staub ◽  
Ewa Szczurek

Computational models for drug sensitivity prediction have the potential to revolutionise personalized cancer medicine. Drug sensitivity assays, as well as profiling of cancer cell lines and drugs becomes increasingly available for training such models. Machine learning methods for drug sensitivity prediction must be optimized for: (i) leveraging the wealth of information about both cancer cell lines and drugs, (ii) predictive performance and (iii) interpretability. Multiple methods were proposed for predicting drug sensitivity from cancer cell line features, some in a multi-task fashion. So far, no such model leveraged drug inhibition profiles. Recent neural network-based recommender systems arise as models capable of predicting cancer cell line response to drugs from their biological features with high prediction accuracy. These models, however, require a tailored approach to model interpretability. In this work, we develop a neural network recommender system for kinase inhibitor sensitivity prediction called DEERS. The model utilizes molecular features of the cancer cell lines and kinase inhibition profiles of the drugs. DEERS incorporates two autoencoders to project cell line and drug features into 10-dimensional hidden representations and a feed-forward neural network to combine them into response prediction. We propose a novel model interpretability approach offering the widest possible assessment of the specific genes and biological processes that underlie the action of the drugs on the cell lines. The approach considers also such genes and processes that were not included in the set of modeled features. Our approach outperforms simpler matrix factorization models, achieving R=0.82 correlation between true and predicted response for the unseen cell lines. Using the interpretability analysis, we evaluate correlation of all human genes with each of the hidden cell line dimensions. Subsequently, we identify 67 biological processes associated with these dimensions. Combined with drug response data, these associations point at the processes that drive the cell line sensitivity to particular compounds. Detailed case studies are shown for PHA-793887, XMD14-99 and Dabrafenib. Our framework provides an expressive, multitask neural network model with a custom interpretability approach for inferring underlying biological factors and explaining cancer cell response to drugs.


2020 ◽  
Vol 13 (S11) ◽  
Author(s):  
Khandakar Tanvir Ahmed ◽  
Sunho Park ◽  
Qibing Jiang ◽  
Yunku Yeu ◽  
TaeHyun Hwang ◽  
...  

Abstract Background Drug sensitivity prediction and drug responsive biomarker selection on high-throughput genomic data is a critical step in drug discovery. Many computational methods have been developed to serve this purpose including several deep neural network models. However, the modular relations among genomic features have been largely ignored in these methods. To overcome this limitation, the role of the gene co-expression network on drug sensitivity prediction is investigated in this study. Methods In this paper, we first introduce a network-based method to identify representative features for drug response prediction by using the gene co-expression network. Then, two graph-based neural network models are proposed and both models integrate gene network information directly into neural network for outcome prediction. Next, we present a large-scale comparative study among the proposed network-based methods, canonical prediction algorithms (i.e., Elastic Net, Random Forest, Partial Least Squares Regression, and Support Vector Regression), and deep neural network models for drug sensitivity prediction. All the source code and processed datasets in this study are available at https://github.com/compbiolabucf/drug-sensitivity-prediction. Results In the comparison of different feature selection methods and prediction methods on a non-small cell lung cancer (NSCLC) cell line RNA-seq gene expression dataset with 50 different drug treatments, we found that (1) the network-based feature selection method improves the prediction performance compared to Pearson correlation coefficients; (2) Random Forest outperforms all the other canonical prediction algorithms and deep neural network models; (3) the proposed graph-based neural network models show better prediction performance compared to deep neural network model; (4) the prediction performance is drug dependent and it may relate to the drug’s mechanism of action. Conclusions Network-based feature selection method and prediction models improve the performance of the drug response prediction. The relations between the genomic features are more robust and stable compared to the correlation between each individual genomic feature and the drug response in high dimension and low sample size genomic datasets.


2020 ◽  
Author(s):  
Yi-Ching Tang ◽  
Assaf Gottlieb

ABSTRACTComputational approaches to predict drug sensitivity can promote precision anticancer therapeutics. Generalizable and explainable models are of critical importance for translation to guide personalized treatment and are often overlooked in favor of prediction performance.Here, we propose a pathway-based model for drug sensitivity prediction that integrates chemical structure information with enrichment of cancer signaling pathways across drug-associated genes, gene expression, mutation and copy number variation data to predict drug response on the Genomics of Drug Sensitivity in Cancer (GDSC) dataset. Using a deep neural network, we outperforming state-of-the-art deep learning models, while demonstrating good generalizability a separate dataset of the Cancer Cell Line Encyclopedia (CCLE) as well as provide explainable results, demonstrated through case studies that are in line with current knowledge. Additionally, our pathway-based model achieved a good performance when predicting unseen drugs and cells, with potential utility for drug development and for guiding individualized medicine.


Sign in / Sign up

Export Citation Format

Share Document