scholarly journals Predicting Cancer Drug Response Using a Recommender System

2017 ◽  
Author(s):  
Chayaporn Supahvilai ◽  
Denis Bertrand ◽  
Niranjan Nagarajan

AbstractMotivationAs we move towards an era of precision medicine, the ability to predict patient-specific drug responses in cancer based on molecular information such as gene expression data represents both an opportunity and a challenge. In particular, methods are needed that can accommodate the high-dimensionality of data to learn interpretable models capturing drug response mechanisms, as well as providing robust predictions across datasets.ResultsWe propose a method based on ideas from “recommender systems” (CaDRReS) that predicts cancer drug responses for unseen cell-lines/patients based on learning projections for drugs and cell-lines into a latent “pharmacogenomic” space. Comparisons with other proposed approaches for this problem based on large public datasets (CCLE, GDSC) shows that CaDRReS provides consistently good models and robust predictions even across unseen patient-derived cell-line datasets. Analysis of the pharmacogenomic spaces inferred by CaDRReS also suggests that they can be used to understand drug mechanisms, identify cellular subtypes, and further characterize drug-pathway associations.AvailabilitySource code and datasets are available at https://github.com/CSB5/[email protected] informationSupplementary data are available online.

2021 ◽  
Author(s):  
Sara JC Gosline ◽  
Cristina Tognon ◽  
Michael Nestor ◽  
Sunil Joshi ◽  
Rucha Modak ◽  
...  

Acute Myeloid Leukemia (AML) affects 20,000 patients in the US annually with a five-year survival rate of approximately 25%. One reason for the low survival rate is the high prevalence of clonal evolution that gives rise to heterogeneous sub-populations of leukemia. This genetic heterogeneity is difficult to treat using conventional therapies that are generally based on the detection of a single driving mutation. Thus, the use of molecular signatures, consisting of multiple functionally related transcripts or proteins, in making treatment decisions may overcome this hurdle and provide a more effective way to inform drug treatment protocols. Toward this end, the Beat AML research program prospectively collected genomic and transcriptomic data from over 1000 AML patients and carried out ex vivo drug sensitivity assays to identify signatures that could predict patient-specific drug responses. The Clinical Proteomic Tumor Analysis Consortium is in the process of extending this cohort to collect proteomic and phosphoproteomic measurements from a subset of these patient samples to evaluate the hypothesis that proteomic signatures can robustly predict drug response in AML patients. We sought to examine this hypothesis on a sub-cohort of 38 patient samples from Beat AML with proteomic and drug response data and evaluate our ability to identify proteomic signatures that predict drug response with high accuracy. For this initial analysis we built predictive models of patient drug responses across 26 drugs of interest using the proteomics and phosphproteomics data. We found that proteomics-derived signatures provide an accurate and robust signature of drug response in the AML ex vivo samples, as well as related cell lines, with better performance than those signatures derived from mutations or mRNA expression. Furthermore, we found that in specific drug-resistant cell lines, the proteins in our prognostic signatures represented dysregulated signaling pathways compared to parental cell lines, confirming the role of the proteins in the signatures in drug resistance. In conclusion, this pilot study demonstrates strong promise for proteomics-based patient stratification to predict drug sensitivity in AML.


2019 ◽  
Vol 35 (14) ◽  
pp. i510-i519 ◽  
Author(s):  
Soufiane Mourragui ◽  
Marco Loog ◽  
Mark A van de Wiel ◽  
Marcel J T Reinders ◽  
Lodewyk F A Wessels

Abstract Motivation Cell lines and patient-derived xenografts (PDXs) have been used extensively to understand the molecular underpinnings of cancer. While core biological processes are typically conserved, these models also show important differences compared to human tumors, hampering the translation of findings from pre-clinical models to the human setting. In particular, employing drug response predictors generated on data derived from pre-clinical models to predict patient response remains a challenging task. As very large drug response datasets have been collected for pre-clinical models, and patient drug response data are often lacking, there is an urgent need for methods that efficiently transfer drug response predictors from pre-clinical models to the human setting. Results We show that cell lines and PDXs share common characteristics and processes with human tumors. We quantify this similarity and show that a regression model cannot simply be trained on cell lines or PDXs and then applied on tumors. We developed PRECISE, a novel methodology based on domain adaptation that captures the common information shared amongst pre-clinical models and human tumors in a consensus representation. Employing this representation, we train predictors of drug response on pre-clinical data and apply these predictors to stratify human tumors. We show that the resulting domain-invariant predictors show a small reduction in predictive performance in the pre-clinical domain but, importantly, reliably recover known associations between independent biomarkers and their companion drugs on human tumors. Availability and implementation PRECISE and the scripts for running our experiments are available on our GitHub page (https://github.com/NKI-CCB/PRECISE). Supplementary information Supplementary data are available at Bioinformatics online.


2020 ◽  
Author(s):  
HARIPRIYA HARIKUMAR ◽  
Thomas P Quinn ◽  
Santu Rana ◽  
Sunil Gupta ◽  
Svetha Venkatesh

Abstract Background: The last decade has seen a major increase in the availability of genomic data. This includes expert-curated databases that describe the biological activity of genes, as well as high-throughput assays that measure gene expression in bulk tissue and single cells. Integrating these heterogeneous data sources can generate new hypotheses about biological systems. Our primary objective is to combine population-level drug-response data with patient-level single-cell expression data to predict how any gene will respond to any drug for any patient.Methods: We take 2 approaches to benchmarking a “dual-channel” random walk with restart (RWR) for data integration. First, we evaluate how well RWR can predict known gene functions from single-cell gene co-expression networks. Second, we evaluate how well RWR can predict known drug responses from individual cell networks. We then present two exploratory applications. In the first application, we combine the Gene Ontology database with glioblastoma single cells from 5 individual patients to identify genes whose functions differ between cancers. In the second application, we combine the LINCS drug-response database with the same glioblastoma data to identify genes that may exhibit patient-specific drug responses.Conclusions: Our manuscript introduces two innovations to the integration of heterogeneous biological data. First, we use a “dual-channel” method to predict up-regulation and down-regulation separately. Second, we use individualized single-cell gene co-expression networks to make personalized predictions. These innovations let us predict gene function and drug response for individual patients. Taken together, our work shows promise that single-cell co-expression data could be combined in heterogeneous information networks to facilitate precision medicine.


2017 ◽  
Author(s):  
Bo Wang ◽  
Daniele Ramazzotti ◽  
Luca De Sano ◽  
Junjie Zhu ◽  
Emma Pierson ◽  
...  

AbstractMotivationWe here present SIMLR (Single-cell Interpretation via Multi-kernel LeaRning), an open-source tool that implements a novel framework to learn a cell-to-cell similarity measure from single-cell RNA-seq data. SIMLR can be effectively used to perform tasks such as dimension reduction, clustering, and visualization of heterogeneous populations of cells. SIMLR was benchmarked against state-of-the-art methods for these three tasks on several public datasets, showing it to be scalable and capable of greatly improving clustering performance, as well as providing valuable insights by making the data more interpretable via better a visualization.Availability and ImplementationSIMLR is available on GitHub in both R and MATLAB implementations. Furthermore, it is also available as an R package on [email protected] or [email protected] InformationSupplementary data are available at Bioinformatics online.


2020 ◽  
Author(s):  
Qiao Liu ◽  
Zhiqiang Hu ◽  
Rui Jiang ◽  
Mu Zhou

AbstractMotivationAccurate prediction of cancer drug response (CDR) is challenging due to the uncertainty of drug efficacy and heterogeneity of cancer patients. Strong evidences have implicated the high dependence of CDR on tumor genomic and transcriptomic profiles of individual patients. Precise identification of CDR is crucial in both guiding anti-cancer drug design and understanding cancer biology.ResultsIn this study, we present DeepCDR which integrates multi-omics profiles of cancer cells and explores intrinsic chemical structures of drugs for predicting cancer drug response. Specifically, DeepCDR is a hybrid graph convolutional network consisting of a uniform graph convolutional network (UGCN) and multiple subnetworks. Unlike prior studies modeling hand-crafted features of drugs, DeepCDR automatically learns the latent representation of topological structures among atoms and bonds of drugs. Extensive experiments showed that DeepCDR outperformed state-of-the-art methods in both classification and regression settings under various data settings. We also evaluated the contribution of different types of omics profiles for assessing drug response. Furthermore, we provided an exploratory strategy for identifying potential cancer-associated genes concerning specific cancer types. Our results highlighted the predictive power of DeepCDR and its potential translational value in guiding disease-specific drug design.AvailabilityDeepCDR is freely available at https://github.com/kimmo1019/[email protected]; [email protected] informationSupplementary data are available at Bioinformatics online.


2020 ◽  
Author(s):  
Evanthia Koukouli ◽  
Dennis Wang ◽  
Frank Dondelinger ◽  
Juhyun Park

AbstractCancer treatments can be highly toxic and frequently only a subset of the patient population will benefit from a given treatment. Tumour genetic makeup plays an important role in cancer drug sensitivity. We suspect that gene expression markers could be used as a decision aid for treatment selection or dosage tuning. Using in vitro cancer cell line dose-response and gene expression data from the Genomics of Drug Sensitivity in Cancer (GDSC) project, we build a dose-varying regression model. Unlike existing approaches, this allows us to estimate dosage-dependent associations with gene expression. We include the transcriptomic profiles as dose-invariant covariates into the regression model and assume that their effect varies smoothly over the dosage levels. A two-stage variable selection algorithm (variable screening followed by penalised regression) is used to identify genetic factors that are associated with drug response over the varying dosages. We evaluate the effectiveness of our method using simulation studies focusing on the choice of tuning parameters and cross-validation for predictive accuracy assessment. We further apply the model to data from five BRAF targeted compounds applied to different cancer cell lines under different dosage levels. We highlight the dosage-dependent dynamics of the associations between the selected genes and drug response, and we perform pathway enrichment analysis to show that the selected genes play an important role in pathways related to tumourgenesis and DNA damage response.Author SummaryTumour cell lines allow scientists to test anticancer drugs in a laboratory environment. Cells are exposed to the drug in increasing concentrations, and the drug response, or amount of surviving cells, is measured. Generally, drug response is summarized via a single number such as the concentration at which 50% of the cells have died (IC50). To avoid relying on such summary measures, we adopted a functional regression approach that takes the dose-response curves as inputs, and uses them to find biomarkers of drug response. One major advantage of our approach is that it describes how the effect of a biomarker on the drug response changes with the drug dosage. This is useful for determining optimal treatment dosages and predicting drug response curves for unseen drug-cell line combinations. Our method scales to large numbers of biomarkers by using regularisation and, in contrast with existing literature, selects the most informative genes by accounting for drug response at untested dosages. We demonstrate its value using data from the Genomics of Drug Sensitivity in Cancer project to identify genes whose expression is associated with drug response. We show that the selected genes recapitulate prior biological knowledge, and belong to known cancer pathways.


2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Haripriya Harikumar ◽  
Thomas P. Quinn ◽  
Santu Rana ◽  
Sunil Gupta ◽  
Svetha Venkatesh

Abstract Background The last decade has seen a major increase in the availability of genomic data. This includes expert-curated databases that describe the biological activity of genes, as well as high-throughput assays that measure gene expression in bulk tissue and single cells. Integrating these heterogeneous data sources can generate new hypotheses about biological systems. Our primary objective is to combine population-level drug-response data with patient-level single-cell expression data to predict how any gene will respond to any drug for any patient. Methods We take 2 approaches to benchmarking a “dual-channel” random walk with restart (RWR) for data integration. First, we evaluate how well RWR can predict known gene functions from single-cell gene co-expression networks. Second, we evaluate how well RWR can predict known drug responses from individual cell networks. We then present two exploratory applications. In the first application, we combine the Gene Ontology database with glioblastoma single cells from 5 individual patients to identify genes whose functions differ between cancers. In the second application, we combine the LINCS drug-response database with the same glioblastoma data to identify genes that may exhibit patient-specific drug responses. Conclusions Our manuscript introduces two innovations to the integration of heterogeneous biological data. First, we use a “dual-channel” method to predict up-regulation and down-regulation separately. Second, we use individualized single-cell gene co-expression networks to make personalized predictions. These innovations let us predict gene function and drug response for individual patients. Taken together, our work shows promise that single-cell co-expression data could be combined in heterogeneous information networks to facilitate precision medicine.


2021 ◽  
Author(s):  
Junyi Chen ◽  
Ren Qi ◽  
Zhenyu Wu ◽  
Anjun Ma ◽  
Lang Li ◽  
...  

Massively bulk RNA sequencing databases incorporating drug screening have opened up an avenue to inform the optimal clinical application of cancer drugs. Meanwhile, the growing single-cell RNA sequencing data contributes to improving therapeutic effectiveness by studying the heterogeneity of drug responses for cancer cell subpopulations. Yet, the drug response information for single-cell data is scarcely obtained. Thus, there is an urgent need to develop computational pipelines to infer and interpret cancer drug responses in single cells. Here, we developed scDEAL, a deep transfer learning framework integrating large-scale bulk and single-cell RNA sequencing drug response datasets. We benchmarked scDEAL on six single-cell RNA sequencing datasets and indicate its model interpretability by several case studies. scDEAL not only achieves accurate and robust performance in single-cell drug response predictions, but also can infer signature genes to reveal potential drug resistance mechanisms based on integrated gradient feature interpretation. This work may help study cell reprogramming, drug selection, and repurposing for improving therapeutic efficacy.


2019 ◽  
Author(s):  
Soufiane Mourragui ◽  
Marco Loog ◽  
Marcel JT Reinders ◽  
Lodewyk FA Wessels

AbstractMotivationCell lines and patient-derived xenografts (PDX) have been used extensively to understand the molecular underpinnings of cancer. While core biological processes are typically conserved, these models also show important differences compared to human tumors, hampering the translation of findings from pre-clinical models to the human setting. In particular, employing drug response predictors generated on data derived from pre-clinical models to predict patient response, remains a challenging task. As very large drug response datasets have been collected for pre-clinical models, and patient drug response data is often lacking, there is an urgent need for methods that efficiently transfer drug response predictors from pre-clinical models to the human setting.ResultsWe show that cell lines and PDXs share common characteristics and processes with human tumors. We quantify this similarity and show that a regression model cannot simply be trained on cell lines or PDXs and then applied on tumors. We developed PRECISE, a novel methodology based on domain adaptation that captures the common information shared amongst pre-clinical models and human tumors in a consensus representation. Employing this representation, we train predictors of drug response on pre-clinical data and apply these predictors to stratify human tumors. We show that the resulting domain-invariant predictors show a small reduction in predictive performance in the pre-clinical domain but, importantly, reliably recover known associations between independent biomarkers and their companion drugs on human tumors.AvailabilityPRECISE and the scripts for running our experiments are available on our GitHub page (https://github.com/NKI-CCB/PRECISE)[email protected] informationSupplementary data are available. online.


2017 ◽  
Author(s):  
Maryam Pouryahya ◽  
Jung Hun Oh ◽  
James Mathews ◽  
Joseph O. Deasy ◽  
Allen Tannenbaum

In the present work, we consider a geometric network approach to study common biological features of anticancer drug response. We use for this purpose the panel of 60 human cell lines (NCI-60) provided by the National Cancer Institute. Our study suggests that utilization of mathematical tools for network-based analysis can provide novel insights into drug response and cancer biology. We adopted a discrete notion of Ricci curvature to measure the robustness of biological networks constructed with a pre-treatment gene expression dataset and coupled the results with the GI50 response of the cell lines to the drugs. The link between network robustness and Ricci curvature was implemented using the theory of optimal mass transport. Our hypothesis behind this idea is that robustness in the biological network contributes to tumor drug resistance, thereby enabling us to predict the effectiveness and sensitivity of drugs in the cell lines. Based on the resulting drug response ranking, we assessed the impact of genes that are likely associated with individual drug response. For important genes identified, we performed a gene ontology enrichment analysis using a curated bioinformatics database which resulted in very plausible biological processes associated with drug response across cell lines and cell types from the biological and literature viewpoint. These results demonstrate the potential of using the mathematical network analysis in assessing drug response and in identifying relevant genomic biomarkers and biological processes for precision medicine.


Sign in / Sign up

Export Citation Format

Share Document