scholarly journals Improving Drug Response Prediction Using Dual Similarity Regularization

2021 ◽  
Author(s):  
Ali Reza Ebadi ◽  
Ali Soleimani ◽  
Abdulbaghi Ghaderzadeh

Abstract Anti-cancer medicine for a particular patient has been a personal medical goal. Many computational models have been proposed by researchers to predict drug response. But predictive accuracy still remains a challenge. Base on this concept which “Similar cells have similar responses to drugs”, we developed the basic method of matrix factorization method by adding fines to similarity. So that the distance of latent factors to two cell lines or (drug) should be inversely related to similarity. This means that two similar drugs or similar cell lines should have a short distance, whereas two similar cell lines or non-similar drugs should have a large gap with their latent factors. We proposed a Dual similarity-regularized matrix factorization (DSRMF) model, then generated new data for drug similarity from the two-dimensional three-dimensional chemical structure, which were obtained from the CCLE and GDSC databases. In this research, by using the proposed model, and generating new drug similarity data we achieved the average Pearson correlation coefficient (PCC) about 0.96, and average mean square error (RMSE) Root about 0.30, between the observed value and the predicted value for the cell line response to the drug. Our analysis in this research showed, using heterogeneous data, has better results, and can be obtained with the proposed model, using other panels’ cancer cell lines, to calculate similarity between cells. Also, by imposing more restrictions on the similarity between cells, we were able to achieve more accurate prediction for the response of the cell line to the anticancer drug.

2020 ◽  
Author(s):  
ALI REZA EBADI ◽  
Ali Soleimani ◽  
ABDULBAGHI GHADERZADEH

Abstract Background:Anti-cancer medicine for a particular patient has been a personal medical goal. Many computational models have been proposed by researchers to predict drug response. But predictive accuracy still remains a challenge. Base on this concept which “Similar cells have similar responses to drugs”, we developed the basic method of matrix factorization method by adding fines to similarity. So that the distance of latent factors to two cell lines or (drug) should be inversely related to similarity. This means that two similar drugs or similar cell lines should have a short distance, whereas two similar cell lines or non-similar drugs should have a large gap with their latent factors.Results:We proposed a Dual similarity-regularized matrix factorization (DSRMF) model, then generated new data for drug similarity from the two-dimensional three-dimensional chemical structure, which were obtained from the CCLE and GDSC databases. In this research, by using the proposed model, and generating new drug similarity data we achieved the average Pearson correlation coefficient (PCC) about 0.96, and average mean square error (RMSE) Root about 0.30, between the observed value and the predicted value for the cell line response to the drug, Conclusions:Our analysis in this research showed, using heterogeneous data, has better results, and can be obtained with the proposed model, using other panels’ cancer cell lines, to calculate similarity between cells. Also, by imposing more restrictions on the similarity between cells, we were able to achieve more accurate prediction for the response of the cell line to the anticancer drug.


Author(s):  
Akram Emdadi ◽  
Changiz Eslahchi

Predicting tumor drug response using cancer cell line drug response values for a large number of anti-cancer drugs is a significant challenge in personalized medicine. Predicting patient response to drugs from data obtained from preclinical models is made easier by the availability of different knowledge on cell lines and drugs. This paper proposes the TCLMF method, a predictive model for predicting drug response in tumor samples that was trained on preclinical samples and is based on the logistic matrix factorization approach. The TCLMF model is designed based on gene expression profiles, tissue type information, the chemical structure of drugs and drug sensitivity (IC 50) data from cancer cell lines. We use preclinical data from the Genomics of Drug Sensitivity in Cancer dataset (GDSC) to train the proposed drug response model, which we then use to predict drug sensitivity of samples from the Cancer Genome Atlas (TCGA) dataset. The TCLMF approach focuses on identifying successful features of cell lines and drugs in order to calculate the probability of the tumor samples being sensitive to drugs. The closest cell line neighbours for each tumor sample are calculated using a description of similarity between tumor samples and cell lines in this study. The drug response for a new tumor is then calculated by averaging the low-rank features obtained from its neighboring cell lines. We compare the results of the TCLMF model with the results of the previously proposed methods using two databases and two approaches to test the model’s performance. In the first approach, 12 drugs with enough known clinical drug response, considered in previous methods, are studied. For 7 drugs out of 12, the TCLMF can significantly distinguish between patients that are resistance to these drugs and the patients that are sensitive to them. These approaches are converted to classification models using a threshold in the second approach, and the results are compared. The results demonstrate that the TCLMF method provides accurate predictions across the results of the other algorithms. Finally, we accurately classify tumor tissue type using the latent vectors obtained from TCLMF’s logistic matrix factorization process. These findings demonstrate that the TCLMF approach produces effective latent vectors for tumor samples. The source code of the TCLMF method is available in https://github.com/emdadi/TCLMF.


BMC Cancer ◽  
2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Shujun Huang ◽  
Pingzhao Hu ◽  
Ted M. Lakowski

Abstract Background Predicting patient drug response based on a patient’s molecular profile is one of the key goals of precision medicine in breast cancer (BC). Multiple drug response prediction models have been developed to address this problem. However, most of them were developed to make sensitivity predictions for multiple single drugs within cell lines from various cancer types instead of a single cancer type, do not take into account drug properties, and have not been validated in cancer patient-derived data. Among the multi-omics data, gene expression profiles have been shown to be the most informative data for drug response prediction. However, these models were often developed with individual genes. Therefore, this study aimed to develop a drug response prediction model for BC using multiple data types from both cell lines and drugs. Methods We first collected the baseline gene expression profiles of 49 BC cell lines along with IC50 values for 220 drugs tested in these cell lines from Genomics of Drug Sensitivity in Cancer (GDSC). Using these data, we developed a multiple-layer cell line-drug response network (ML-CDN2) by integrating a one-layer cell line similarity network based on the pathway activity profiles and a three-layer drug similarity network based on the drug structures, targets, and pan-cancer IC50 profiles. We further used ML-CDN2 to predict the drug response for new BC cell lines or patient-derived samples. Results ML-CDN2 demonstrated a good predictive performance, with the Pearson correlation coefficient between the observed and predicted IC50 values for all GDSC cell line-drug pairs of 0.873. Also, ML-CDN2 showed a good performance when used to predict drug response in new BC cell lines from the Cancer Cell Line Encyclopedia (CCLE), with a Pearson correlation coefficient of 0.718. Moreover, we found that the cell line-derived ML-CDN2 model could be applied to predict drug response in the BC patient-derived samples from The Cancer Genome Atlas (TCGA). Conclusions The ML-CDN2 model was built to predict BC drug response using comprehensive information from both cell lines and drugs. Compared with existing methods, it has the potential to predict the drug response for BC patient-derived samples.


2021 ◽  
Vol 22 (1) ◽  
Author(s):  
Akram Emdadi ◽  
Changiz Eslahchi

Abstract Background Predicting the response of cancer cell lines to specific drugs is an essential problem in personalized medicine. Since drug response is closely associated with genomic information in cancer cells, some large panels of several hundred human cancer cell lines are organized with genomic and pharmacogenomic data. Although several methods have been developed to predict the drug response, there are many challenges in achieving accurate predictions. This study proposes a novel feature selection-based method, named Auto-HMM-LMF, to predict cell line-drug associations accurately. Because of the vast dimensions of the feature space for predicting the drug response, Auto-HMM-LMF focuses on the feature selection issue for exploiting a subset of inputs with a significant contribution. Results This research introduces a novel method for feature selection of mutation data based on signature assignments and hidden Markov models. Also, we use the autoencoder models for feature selection of gene expression and copy number variation data. After selecting features, the logistic matrix factorization model is applied to predict drug response values. Besides, by comparing to one of the most powerful feature selection methods, the ensemble feature selection method (EFS), we showed that the performance of the predictive model based on selected features introduced in this paper is much better for drug response prediction. Two datasets, the Genomics of Drug Sensitivity in Cancer (GDSC) and Cancer Cell Line Encyclopedia (CCLE) are used to indicate the efficiency of the proposed method across unseen patient cell-line. Evaluation of the proposed model showed that Auto-HMM-LMF could improve the accuracy of the results of the state-of-the-art algorithms, and it can find useful features for the logistic matrix factorization method. Conclusions We depicted an application of Auto-HMM-LMF in exploring the new candidate drugs for head and neck cancer that showed the proposed method is useful in drug repositioning and personalized medicine. The source code of Auto-HMM-LMF method is available in https://github.com/emdadi/Auto-HMM-LMF.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1623-1623 ◽  
Author(s):  
Karen Dybkær ◽  
Hanne Due ◽  
Rasmus Froberg Brøndum ◽  
Ken H. Young ◽  
Martin Bøgsted

Background: Patients with Diffuse large B-cell lymphoma (DLBCL) in approximately 40% of cases suffer from primary refractory disease and treatment induced immuno-chemotherapy resistance demonstrating that standard provided treatment regimens are not sufficient to cure all patients. Early detection of resistance is of great importance and defining microRNA (miRNA) involvement in resistance could be useful to guide treatment selection and help monitor treatment administration while sparing patients for inefficient, but still toxic therapy. Concept and Aims: With information on drug-response specific miRNAs, we hypothesized that multi-miRNA panels can improve robustness of individual clinical markers and serve as a prognostic classifier predicting disease progression in DLBCL patients. Methods: Fifteen DLBCL cell lines were tested for sensitivity towards rituximab (R), cyclophosphamide (C), doxorubicin (H), and vincristine (O). Cell line specific seeding concentrations was used to ensure exponential growth and each cell line was subjected to 16 concentrations in serial 2-fold dilutions and number of metabolic active cells was evaluated after 48 hours of drug exposure using MTS assay. For each drug, we ranked the cell lines according to their sensitivity and categorized them as sensitive, intermediate responsive, or resistant. Differential miRNA expression analysis between sensitive and resistant cell lines identified 43 miRNAs to be associated with response to compounds of the R-CHOP regimen, by selecting probes with a log fold change larger than 2. Baseline miRNA expression data were obtained for each cell line in untreated condition, and differential miRNA expression analysis identified 43 miRNAs associated to response to R-CHOP. Using the Affymetrix HG-U133+2 platform, expression levels of the miRNA precursors were assessed in 701 diagnostic DLBCL biopsies, and miRNA-panel classifiers were build using multiple Cox regression or random survival forest. Results: Generated prognostic miRNA-panel classifiers were tested for predictive accuracies and were subsequently evaluated by Brier scores and time varying area under the ROC curves (tAUC). Progression-free survival (PFS) was chosen as the outcome, since it is a treatment evaluation parameter as closely as possible to the time of drug exposure and the tested miRNAs were all associated directly to drug specific response. Furthermore, overall survival (OS) was used for verification of findings. Comparison of analyses conducted for the respective cohorts (All DLBCL, ABC, and GCB patients) showed the lowest prediction errors for all models within the GCB subclass with a multivariate Cox miRNA-panel model including miR-146a, miR-155, miR-21, miR-34a, and miR-23a~miR-27a~miR-24-2 cluster performed the best and successfully stratified GCB-DLBCL patients into high- and low-risk of disease progression. In addition, combination of the miRNA-panel and international prognostic index (IPI) substantially increased prognostic performance in GCB classified patients, indicating a prognostic signal from the response-specific miRNAs independent of IPI. In conclusion: We found as proof of concept that adding gene expression data detecting drug-response specific miRNAs to the clinically established IPI improved the prognostic stratification of GCB-DLBCL patients treated with R-CHOP. Disclosures No relevant conflicts of interest to declare.


2013 ◽  
Vol 31 (15_suppl) ◽  
pp. e14544-e14544
Author(s):  
Eva Budinska ◽  
Jenny Wilding ◽  
Vlad Calin Popovici ◽  
Edoardo Missiaglia ◽  
Arnaud Roth ◽  
...  

e14544 Background: We identified CRC gene expression subtypes (ASCO 2012, #3511), which associate with established parameters of outcome as well as relevant biological motifs. We now substantiate their biological and potentially clinical significance by linking them with cell line data and drug sensitivity, primarily attempting to identify models for the poor prognosis subtypes Mesenchymal and CIMP-H like (characterized by EMT/stroma and immune-associated gene modules, respectively). Methods: We analyzed gene expression profiles of 35 publicly available cell lines with sensitivity data for 82 drug compounds, and our 94 cell lines with data on sensitivity for 7 compounds and colony morphology. As in vitro, stromal and immune-associated genes loose their relevance, we trained a new classifier based on genes expressed in both systems, which identifies the subtypes in both tissue and cell cultures. Cell line subtypes were validated by comparing their enrichment for molecular markers with that of our CRC subtypes. Drug sensitivity was assessed by linking original subtypes with 92 drug response signatures (MsigDB) via gene set enrichment analysis, and by screening drug sensitivity of cell line panels against our subtypes (Kruskal-Wallis test). Results: Of the cell lines 70% could be assigned to a subtype with a probability as high as 0.95. The cell line subtypes were significantly associated with their KRAS, BRAF and MSI status and corresponded to our CRC subtypes. Interestingly, the cell lines which in matrigel created a network of undifferentiated cells were assigned to the Mesenchymal subtype. Drug response studies revealed potential sensitivity of subtypes to multiple compounds, in addition to what could be predicted based on their mutational profile (e.g. sensitivity of the CIMP-H subtype to Dasatinib, p<0.01). Conclusions: Our data support the biological and potentially clinical significance of the CRC subtypes in their association with cell line models, including results of drug sensitivity analysis. Our subtypes might not only have prognostic value but might also be predictive for response to drugs. Subtyping cell lines further substantiates their significance as relevant model for functional studies.


2021 ◽  
Author(s):  
Sara Pidò ◽  
Carolina Testa ◽  
Pietro Pinoli

AbstractLarge annotated cell line collections have been proven to enable the prediction of drug response in the preclinical setting. We present an enhancement of Non-Negative Matrix Tri-Factorization method, which allows the integration of different data types for the prediction of missing associations. To test our method we retrieved a dataset from CCLE, containing the connections among cell lines and drugs by means of their IC50 values. We performed two different kind of experiments: a) prediction of missing values in the matrix, b) prediction of the complete drug profile of a new cell line, demonstrating the validity of the method in both scenarios.


2019 ◽  
Vol 17 ◽  
pp. 164-174 ◽  
Author(s):  
Na-Na Guan ◽  
Yan Zhao ◽  
Chun-Chun Wang ◽  
Jian-Qiang Li ◽  
Xing Chen ◽  
...  

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.


2019 ◽  
Vol 10 (1) ◽  
Author(s):  
Yongsoo Kim ◽  
Tycho Bismeijer ◽  
Wilbert Zwart ◽  
Lodewyk F. A. Wessels ◽  
Daniel J. Vis

Abstract Integrative analyses that summarize and link molecular data to treatment sensitivity are crucial to capture the biological complexity which is essential to further precision medicine. We introduce Weighted Orthogonal Nonnegative parallel factor analysis (WON-PARAFAC), a data integration method that identifies sparse and interpretable factors. WON-PARAFAC summarizes the GDSC1000 cell line compendium in 130 factors. We interpret the factors based on their association with recurrent molecular alterations, pathway enrichment, cancer type, and drug-response. Crucially, the cell line derived factors capture the majority of the relevant biological variation in Patient-Derived Xenograft (PDX) models, strongly suggesting our factors capture invariant and generalizable aspects of cancer biology. Furthermore, drug response in cell lines is better and more consistently translated to PDXs using factor-based predictors as compared to raw feature-based predictors. WON-PARAFAC efficiently summarizes and integrates multiway high-dimensional genomic data and enhances translatability of drug response prediction from cell lines to patient-derived xenografts.


Sign in / Sign up

Export Citation Format

Share Document