Deep learning for drug response prediction in cancer

Author(s):  
Delora Baptista ◽  
Pedro G Ferreira ◽  
Miguel Rocha

Abstract Predicting the sensitivity of tumors to specific anti-cancer treatments is a challenge of paramount importance for precision medicine. Machine learning(ML) algorithms can be trained on high-throughput screening data to develop models that are able to predict the response of cancer cell lines and patients to novel drugs or drug combinations. Deep learning (DL) refers to a distinct class of ML algorithms that have achieved top-level performance in a variety of fields, including drug discovery. These types of models have unique characteristics that may make them more suitable for the complex task of modeling drug response based on both biological and chemical data, but the application of DL to drug response prediction has been unexplored until very recently. The few studies that have been published have shown promising results, and the use of DL for drug response prediction is beginning to attract greater interest from researchers in the field. In this article, we critically review recently published studies that have employed DL methods to predict drug response in cancer cell lines. We also provide a brief description of DL and the main types of architectures that have been used in these studies. Additionally, we present a selection of publicly available drug screening data resources that can be used to develop drug response prediction models. Finally, we also address the limitations of these approaches and provide a discussion on possible paths for further improvement. Contact:[email protected]

Genes ◽  
2021 ◽  
Vol 12 (6) ◽  
pp. 844
Author(s):  
Abhishek Majumdar ◽  
Yueze Liu ◽  
Yaoqin Lu ◽  
Shaofeng Wu ◽  
Lijun Cheng

Background: Cancer cell lines are frequently used in research as in-vitro tumor models. Genomic data and large-scale drug screening have accelerated the right drug selection for cancer patients. Accuracy in drug response prediction is crucial for success. Due to data-type diversity and big data volume, few methods can integrative and efficiently find the principal low-dimensional manifold of the high-dimensional cancer multi-omics data to predict drug response in precision medicine. Method: A novelty k-means Ensemble Support Vector Regression (kESVR) is developed to predict each drug response values for single patient based on cell-line gene expression data. The kESVR is a blend of supervised and unsupervised learning methods and is entirely data driven. It utilizes embedded clustering (Principal Component Analysis and k-means clustering) and local regression (Support Vector Regression) to predict drug response and obtain the global pattern while overcoming missing data and outliers’ noise. Results: We compared the efficiency and accuracy of kESVR to 4 standard machine learning regression models: (1) simple linear regression, (2) support vector regression (3) random forest (quantile regression forest) and (4) back propagation neural network. Our results, which based on drug response across 610 cancer cells from Cancer Cell Line Encyclopedia (CCLE) and Cancer Therapeutics Response Portal (CTRP v2), proved to have the highest accuracy (smallest mean squared error (MSE) measure). We next compared kESVR with existing 17 drug response prediction models based a varied range of methods such as regression, Bayesian inference, matrix factorization and deep learning. After ranking the 18 models based on their accuracy of prediction, kESVR ranks first (best performing) in majority (74%) of the time. As for the remaining (26%) cases, kESVR still ranked in the top five performing models. Conclusion: In this paper we introduce a novel model (kESVR) for drug response prediction using high dimensional cell-line gene expression data. This model outperforms current existing prediction models in terms of prediction accuracy and speed and overcomes overfitting. This can be used in future to develop a robust drug response prediction system for cancer patients using the cancer cell-lines guidance and multi-omics data.


2018 ◽  
Vol 8 (1) ◽  
Author(s):  
Fei Zhang ◽  
Minghui Wang ◽  
Jianing Xi ◽  
Jianghong Yang ◽  
Ao Li

2020 ◽  
Vol 16 (1) ◽  
pp. 31-38
Author(s):  
Shiming Wang ◽  
Jie Li

Drug response prediction in cancer cell lines is vital to discover anticancer drugs for new cell lines.


2018 ◽  
Vol 35 (9) ◽  
pp. 1527-1535 ◽  
Author(s):  
Jianghong Yang ◽  
Ao Li ◽  
Yongqiang Li ◽  
Xiangqian Guo ◽  
Minghui Wang

2021 ◽  
Author(s):  
David Earl Hostallero ◽  
Lixuan Wei ◽  
Liewei Wang ◽  
Junmei Cairns ◽  
Amin Emad

Background: Prediction of the response of cancer patients to different treatments and identification of biomarkers of drug sensitivity are two major goals of individualized medicine. In this study, we developed a deep learning framework called TINDL, completely trained on preclinical cancer cell lines, to predict the response of cancer patients to different treatments. TINDL utilizes a tissue-informed normalization to account for the tissue and cancer type of the tumours and to reduce the statistical discrepancies between cell lines and patient tumours. In addition, this model identifies a small set of genes whose mRNA expression are predictive of drug response in the trained model, enabling identification of biomarkers of drug sensitivity. Results: Using data from two large databases of cancer cell lines and cancer tumours, we showed that this model can distinguish between sensitive and resistant tumours for 10 (out of 14) drugs, outperforming various other machine learning models. In addition, our siRNA knockdown experiments on 10 genes identified by this model for one of the drugs (tamoxifen) confirmed that all of these genes significantly influence the drug sensitivity of the MCF7 cell line to this drug. In addition, genes implicated for multiple drugs pointed to shared mechanism of action among drugs and suggested several important signaling pathways. Conclusions: In summary, this study provides a powerful deep learning framework for prediction of drug response and for identification of biomarkers of drug sensitivity in cancer.


2016 ◽  
Vol 11 (2) ◽  
pp. 203-210 ◽  
Author(s):  
Jiguang Wang ◽  
Judith Kribelbauer ◽  
Raul Rabadan

2019 ◽  
Vol 9 (1) ◽  
Author(s):  
Farnoosh Abbas-Aghababazadeh ◽  
Pengcheng Lu ◽  
Brooke L. Fridley

Abstract Cancer cell lines (CCLs) have been widely used to study of cancer. Recent studies have called into question the reliability of data collected on CCLs. Hence, we set out to determine CCLs that tend to be overly sensitive or resistant to a majority of drugs utilizing a nonlinear mixed-effects (NLME) modeling framework. Using drug response data collected in the Cancer Cell Line Encyclopedia (CCLE) and the Genomics of Drug Sensitivity in Cancer (GDSC), we determined the optimal functional form for each drug. Then, a NLME model was fit to the drug response data, with the estimated random effects used to determine sensitive or resistant CCLs. Out of the roughly 500 CCLs studies from the CCLE, we found 17 cell lines to be overly sensitive or resistant to the studied drugs. In the GDSC, we found 15 out of the 990 CCLs to be excessively sensitive or resistant. These results can inform researchers in the selection of CCLs to include in drug studies. Additionally, this study illustrates the need for assessing the dose-response functional form and the use of NLME models to achieve more stable estimates of drug response parameters.


Sign in / Sign up

Export Citation Format

Share Document