1
|
Xie X, Wang F, Wang G, Zhu W, Du X, Wang H. Learning the cellular activity representation based on gene regulatory networks for prediction of tumor response to drugs. Artif Intell Med 2024; 152:102864. [PMID: 38640702 DOI: 10.1016/j.artmed.2024.102864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 01/28/2024] [Accepted: 03/30/2024] [Indexed: 04/21/2024]
Abstract
Predicting the response of tumor cells to anti-tumor drugs is critical to realizing cancer precision medicine. Currently, most existing methods ignore the regulatory relationships between genes and thus have unsatisfactory predictive performance. In this paper, we propose to predict anti-tumor drug efficacy via learning the activity representation of tumor cells based on a priori knowledge of gene regulation networks (GRNs). Specifically, the method simulates the cellular biosystem by synthesizing a cell-gene activity network and then infers a new low-dimensional activity representation for tumor cells from the raw high-dimensional expression profile. The simulated cell-gene network mainly comprises known gene regulatory networks collected from multiple resources and fuses tumor cells by linking them to hotspot genes that are over- or under-expressed in them. The resulting activity representation could not only reflect the shallow expression profile (hotspot genes) but also mines in-depth information of gene regulation activity in tumor cells before treatment. Finally, we build deep learning models on the activity representation for predicting drug efficacy in tumor cells. Experimental results on the benchmark GDSC dataset demonstrate the superior performance of the proposed method over SOTA methods with the highest AUC of 0.954 in the efficacy label prediction and the best R2 of 0.834 in the regression of half maximal inhibitory concentration (IC50) values, suggesting the potential value of the proposed method in practice.
Collapse
Affiliation(s)
- Xinping Xie
- School of mathematics and physics, Anhui Jianzhu University, Hefei, China
| | - Fengting Wang
- School of mathematics and physics, Anhui Jianzhu University, Hefei, China; Institute of Intelligent Machines, Hefei Institutes of Physical Science, CAS, Hefei, China
| | - Guanfu Wang
- School of mathematics and physics, Anhui Jianzhu University, Hefei, China
| | - Weiwei Zhu
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, CAS, Hefei, China; Zhongqi AI Lab, Hefei, China
| | - Xiaodong Du
- Experimental Teaching Center, Hefei University, Hefei, China
| | - Hongqiang Wang
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, CAS, Hefei, China; Zhongqi AI Lab, Hefei, China.
| |
Collapse
|
2
|
Narykov O, Zhu Y, Brettin T, Evrard YA, Partin A, Shukla M, Xia F, Clyde A, Vasanthakumari P, Doroshow JH, Stevens RL. Integration of Computational Docking into Anti-Cancer Drug Response Prediction Models. Cancers (Basel) 2023; 16:50. [PMID: 38201477 PMCID: PMC10777918 DOI: 10.3390/cancers16010050] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 01/12/2024] Open
Abstract
Cancer is a heterogeneous disease in that tumors of the same histology type can respond differently to a treatment. Anti-cancer drug response prediction is of paramount importance for both drug development and patient treatment design. Although various computational methods and data have been used to develop drug response prediction models, it remains a challenging problem due to the complexities of cancer mechanisms and cancer-drug interactions. To better characterize the interaction between cancer and drugs, we investigate the feasibility of integrating computationally derived features of molecular mechanisms of action into prediction models. Specifically, we add docking scores of drug molecules and target proteins in combination with cancer gene expressions and molecular drug descriptors for building response models. The results demonstrate a marginal improvement in drug response prediction performance when adding docking scores as additional features, through tests on large drug screening data. We discuss the limitations of the current approach and provide the research community with a baseline dataset of the large-scale computational docking for anti-cancer drugs.
Collapse
Affiliation(s)
- Oleksandr Narykov
- Computing, Environment and Life Sciences, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (T.B.); (A.P.); (M.S.); (F.X.); (P.V.); (R.L.S.)
| | - Yitan Zhu
- Computing, Environment and Life Sciences, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (T.B.); (A.P.); (M.S.); (F.X.); (P.V.); (R.L.S.)
| | - Thomas Brettin
- Computing, Environment and Life Sciences, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (T.B.); (A.P.); (M.S.); (F.X.); (P.V.); (R.L.S.)
| | - Yvonne A. Evrard
- Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA;
| | - Alexander Partin
- Computing, Environment and Life Sciences, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (T.B.); (A.P.); (M.S.); (F.X.); (P.V.); (R.L.S.)
| | - Maulik Shukla
- Computing, Environment and Life Sciences, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (T.B.); (A.P.); (M.S.); (F.X.); (P.V.); (R.L.S.)
| | - Fangfang Xia
- Computing, Environment and Life Sciences, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (T.B.); (A.P.); (M.S.); (F.X.); (P.V.); (R.L.S.)
| | - Austin Clyde
- Computing, Environment and Life Sciences, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (T.B.); (A.P.); (M.S.); (F.X.); (P.V.); (R.L.S.)
- Department of Computer Science, The University of Chicago, Chicago, IL 60637, USA
| | - Priyanka Vasanthakumari
- Computing, Environment and Life Sciences, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (T.B.); (A.P.); (M.S.); (F.X.); (P.V.); (R.L.S.)
| | - James H. Doroshow
- Developmental Therapeutics Branch, National Cancer Institute, Bethesda, MD 20892, USA;
| | - Rick L. Stevens
- Computing, Environment and Life Sciences, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (T.B.); (A.P.); (M.S.); (F.X.); (P.V.); (R.L.S.)
- Department of Computer Science, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
3
|
Liu X, Zhang W. A subcomponent-guided deep learning method for interpretable cancer drug response prediction. PLoS Comput Biol 2023; 19:e1011382. [PMID: 37603576 PMCID: PMC10470940 DOI: 10.1371/journal.pcbi.1011382] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 08/31/2023] [Accepted: 07/24/2023] [Indexed: 08/23/2023] Open
Abstract
Accurate prediction of cancer drug response (CDR) is a longstanding challenge in modern oncology that underpins personalized treatment. Current computational methods implement CDR prediction by modeling responses between entire drugs and cell lines, without the consideration that response outcomes may primarily attribute to a few finer-level 'subcomponents', such as privileged substructures of the drug or gene signatures of the cancer cell, thus producing predictions that are hard to explain. Herein, we present SubCDR, a subcomponent-guided deep learning method for interpretable CDR prediction, to recognize the most relevant subcomponents driving response outcomes. Technically, SubCDR is built upon a line of deep neural networks that enables a set of functional subcomponents to be extracted from each drug and cell line profile, and breaks the CDR prediction down to identifying pairwise interactions between subcomponents. Such a subcomponent interaction form can offer a traceable path to explicitly indicate which subcomponents contribute more to the response outcome. We verify the superiority of SubCDR over state-of-the-art CDR prediction methods through extensive computational experiments on the GDSC dataset. Crucially, we found many predicted cases that demonstrate the strength of SubCDR in finding the key subcomponents driving responses and exploiting these subcomponents to discover new therapeutic drugs. These results suggest that SubCDR will be highly useful for biomedical researchers, particularly in anti-cancer drug design.
Collapse
Affiliation(s)
- Xuan Liu
- College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Wen Zhang
- College of Informatics, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
4
|
Hostallero DE, Wei L, Wang L, Cairns J, Emad A. Preclinical-to-clinical Anti-cancer Drug Response Prediction and Biomarker Identification Using TINDL. GENOMICS, PROTEOMICS & BIOINFORMATICS 2023; 21:535-550. [PMID: 36775056 PMCID: PMC10787192 DOI: 10.1016/j.gpb.2023.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/28/2022] [Accepted: 01/31/2023] [Indexed: 02/12/2023]
Abstract
Prediction of the response of cancer patients to different treatments and identification of biomarkers of drug response are two major goals of individualized medicine. Here, we developed a deep learning framework called TINDL, completely trained on preclinical cancer cell lines (CCLs), to predict the response of cancer patients to different treatments. TINDL utilizes a tissue-informed normalization to account for the tissue type and cancer type of the tumors and to reduce the statistical discrepancies between CCLs and patient tumors. Moreover, by making the deep learning black box interpretable, this model identifies a small set of genes whose expression levels are predictive of drug response in the trained model, enabling identification of biomarkers of drug response. Using data from two large databases of CCLs and cancer tumors, we showed that this model can distinguish between sensitive and resistant tumors for 10 (out of 14) drugs, outperforming various other machine learning models. In addition, our small interfering RNA (siRNA) knockdown experiments on 10 genes identified by this model for one of the drugs (tamoxifen) confirmed that tamoxifen sensitivity is substantially influenced by all of these genes in MCF7 cells, and seven of these genes in T47D cells. Furthermore, genes implicated for multiple drugs pointed to shared mechanism of action among drugs and suggested several important signaling pathways. In summary, this study provides a powerful deep learning framework for prediction of drug response and identification of biomarkers of drug response in cancer. The code can be accessed at https://github.com/ddhostallero/tindl.
Collapse
Affiliation(s)
- David Earl Hostallero
- Department of Electrical and Computer Engineering, McGill University, Montreal, QC H3A, Canada; Mila - Quebec Artificial Intelligence Institute, Montreal, QC H2S, Canada
| | - Lixuan Wei
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Junmei Cairns
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA.
| | - Amin Emad
- Department of Electrical and Computer Engineering, McGill University, Montreal, QC H3A, Canada; Mila - Quebec Artificial Intelligence Institute, Montreal, QC H2S, Canada; The Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A, Canada.
| |
Collapse
|
5
|
Singh DP, Kaushik B. CTDN (Convolutional Temporal Based Deep- Neural Network): An Improvised Stacked Hybrid Computational Approach for Anticancer Drug Response Prediction. Comput Biol Chem 2023; 105:107868. [PMID: 37257399 DOI: 10.1016/j.compbiolchem.2023.107868] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 06/02/2023]
Abstract
The characterization of drug - metabolizing enzymes is a significant problem for customized therapy. It is important to choose the right drugs for cancer victims, and the ability to forecast how those drugs will react is usually based on the available information, genetic sequence, and structural properties. To the finest of our knowledge, this is the first study to evaluate optimization algorithms for selection of features and pharmacogenetics categorization using classification methods based on a successful evolutionary algorithm using datasets from the Cancer Cell Line Encyclopaedia (CCLE) and Genomics of Drug Sensitivity in Cancer (GDSC). The study proposes the uses of Firefly and Grey Wolf Optimization techniques for feature extraction, while comparing the traditional Machine Learning (ML), ensemble ML and Stacking Algorithm with the proposed Convolutional Temporal Deep Neural Network or CTDN. With the potential to increase efficiency from the suggested intelligible classifier model for a suggestive chemotherapeutic drugs response prediction, our study is important in particular for selecting an acceptable feature selection method. The comparison analysis demonstrates that the proposed model not only surpasses the prior state-of-the-art methods, but also uses Grey Wolf and Fire Fly Optimization to lessen multicollinearity and overfitting.
Collapse
Affiliation(s)
- Davinder Paul Singh
- School of Computer Science and Engineering, Shri Mata Vaishno Devi University, Katra 182320, Jammu and Kashmir, India.
| | - Baijnath Kaushik
- School of Computer Science and Engineering, Shri Mata Vaishno Devi University, Katra 182320, Jammu and Kashmir, India
| |
Collapse
|
6
|
Lee K, Cho D, Jang J, Choi K, Jeong HO, Seo J, Jeong WK, Lee S. RAMP: response-aware multi-task learning with contrastive regularization for cancer drug response prediction. Brief Bioinform 2023; 24:6865135. [PMID: 36460623 DOI: 10.1093/bib/bbac504] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 10/13/2022] [Accepted: 10/24/2022] [Indexed: 12/05/2022] Open
Abstract
The accurate prediction of cancer drug sensitivity according to the multiomics profiles of individual patients is crucial for precision cancer medicine. However, the development of prediction models has been challenged by the complex crosstalk of input features and the resistance-dominant drug response information contained in public databases. In this study, we propose a novel multidrug response prediction framework, response-aware multitask prediction (RAMP), via a Bayesian neural network and restrict it by soft-supervised contrastive regularization. To utilize network embedding vectors as representation learning features for heterogeneous networks, we harness response-aware negative sampling, which applies cell line-drug response information to the training of network embeddings. RAMP overcomes the prediction accuracy limitation induced by the imbalance of trained response data based on the comprehensive selection and utilization of drug response features. When trained on the Genomics of Drug Sensitivity in Cancer dataset, RAMP achieved an area under the receiver operating characteristic curve > 89%, an area under the precision-recall curve > 59% and an $\textrm{F}_1$ score > 52% and outperformed previously developed methods on both balanced and imbalanced datasets. Furthermore, RAMP predicted many missing drug responses that were not included in the public databases. Our results showed that RAMP will be suitable for the high-throughput prediction of cancer drug sensitivity and will be useful for guiding cancer drug selection processes. The Python implementation for RAMP is available at https://github.com/hvcl/RAMP.
Collapse
Affiliation(s)
- Kanggeun Lee
- Department of Computer Science and Engineering at Korea University
| | - Dongbin Cho
- Department of Computer Science at Hanyang University
| | - Jinho Jang
- Department of Biomedical Engineering at UNIST
| | - Kang Choi
- Department of Computer Science at Hanyang University
| | | | - Jiwon Seo
- Department of Computer Science at Hanyang University
| | - Won-Ki Jeong
- Department of Computer Science and Engineering at Korea University
| | - Semin Lee
- Department of Biomedical Engineering at UNIST
| |
Collapse
|
7
|
Xi J, Wang D, Yang X, Zhang W, Huang Q. Cancer omic data based explainable AI drug recommendation inference: A traceability perspective for explainability. Biomed Signal Process Control 2023. [DOI: 10.1016/j.bspc.2022.104144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
8
|
Identification of phenocopies improves prediction of targeted therapy response over DNA mutations alone. NPJ Genom Med 2022; 7:58. [PMID: 36253482 PMCID: PMC9576758 DOI: 10.1038/s41525-022-00328-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/29/2022] [Indexed: 11/09/2022] Open
Abstract
DNA mutations in specific genes can confer preferential benefit from drugs targeting those genes. However, other molecular perturbations can “phenocopy” pathogenic mutations, but would not be identified using standard clinical sequencing, leading to missed opportunities for other patients to benefit from targeted treatments. We hypothesized that RNA phenocopy signatures of key cancer driver gene mutations could improve our ability to predict response to targeted therapies, despite not being directly trained on drug response. To test this, we built gene expression signatures in tissue samples for specific mutations and found that phenocopy signatures broadly increased accuracy of drug response predictions in-vitro compared to DNA mutation alone, and identified additional cancer cell lines that respond well with a positive/negative predictive value on par or better than DNA mutations. We further validated our results across four clinical cohorts. Our results suggest that routine RNA sequencing of tumors to identify phenocopies in addition to standard targeted DNA sequencing would improve our ability to accurately select patients for targeted therapies in the clinic.
Collapse
|
9
|
Chen J, Huang J, Liao Y, Zhu L, Cai H. Identify Multiple Gene-Drug Common Modules Via Constrained Graph Matching. IEEE J Biomed Health Inform 2022; 26:4794-4805. [PMID: 35788454 DOI: 10.1109/jbhi.2022.3188503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Identifying gene-drug interactions is vital to understanding biological mechanisms and achieving precise drug repurposing. High-throughput technologies produce a large amount of pharmacological and genomic data, providing an opportunity to explore the associations between oncogenic genes and therapeutic drugs. However, most studies only focus on "one-to-one" or "one-to-many" interactions, ignoring the multivariate patterns between genes and drugs. In this article, a high-order graph matching model with hypergraph constraints is proposed to discover the gene-drug common regulatory modules. Moreover, the prior knowledge is formulated into hypergraph constraints to reveal their multiple correspondences, penalizing the tensor matching process. The experimental results on the synthetic data demonstrate the proposed model is robust to noise contamination and outlier corruption, achieving a better performance than four state-of-the-art methods. We then evaluate the statistical power of our proposed method on the pharmacogenomics data. Our identified gene-drug common modules not only show significantly enriched pathways associated with cancer but also manifest the highly close gene-drug interactions.
Collapse
|
10
|
Hostallero DE, Li Y, Emad A. Looking at the BiG Picture: Incorporating bipartite graphs in drug response prediction. Bioinformatics 2022; 38:3609-3620. [PMID: 35674359 DOI: 10.1093/bioinformatics/btac383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 04/17/2022] [Accepted: 06/01/2022] [Indexed: 12/15/2022] Open
Abstract
MOTIVATION The increasing number of publicly available databases containing drugs' chemical structures, their response in cell lines, and molecular profiles of the cell lines has garnered attention to the problem of drug response prediction. However, many existing methods do not fully leverage the information that is shared among cell lines and drugs with similar structure. As such, drug similarities in terms of cell line responses and chemical structures could prove to be useful in forming drug representations to improve drug response prediction accuracy. RESULTS We present two deep learning approaches, BiG-DRP and BiG-DRP+, for drug response prediction. Our models take advantage of the drugs' chemical structure and the underlying relationships of drugs and cell lines through a bipartite graph and a heterogenous graph convolutional network that incorporate sensitive and resistant cell line information in forming drug representations. Evaluation of our methods and other state-of-the-art models in different scenarios shows that incorporating this bipartite graph significantly improves the prediction performance. Additionally, genes that contribute significantly to the performance of our models also point to important biological processes and signaling pathways. Analysis of predicted drug response of patients' tumors using our model revealed important associations between mutations and drug sensitivity, illustrating the utility of our model in pharmacogenomics studies. AVAILABILITY AND IMPLEMENTATION An implementation of the algorithms in Python is provided in https://github.com/ddhostallero/BiG-DRP. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- David Earl Hostallero
- Department of Electrical and Computer Engineering, McGill University, Montreal, QC H3A 0E9, Canada
- Mila, Quebec AI Institute, Montreal, QC H2S 3H1, Canada
| | - Yihui Li
- Department of Electrical and Computer Engineering, McGill University, Montreal, QC H3A 0E9, Canada
| | - Amin Emad
- Department of Electrical and Computer Engineering, McGill University, Montreal, QC H3A 0E9, Canada
- Mila, Quebec AI Institute, Montreal, QC H2S 3H1, Canada
- The Rosalind and Morris Goodman Cancer Institute, Montreal, QC H3A 1A3, Canada
| |
Collapse
|
11
|
Feng YH, Zhang SW. Prediction of Drug-Drug Interaction Using an Attention-Based Graph Neural Network on Drug Molecular Graphs. Molecules 2022; 27:molecules27093004. [PMID: 35566354 PMCID: PMC9105425 DOI: 10.3390/molecules27093004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 04/28/2022] [Accepted: 04/30/2022] [Indexed: 12/04/2022] Open
Abstract
The treatment of complex diseases by using multiple drugs has become popular. However, drug-drug interactions (DDI) may give rise to the risk of unanticipated adverse effects and even unknown toxicity. Therefore, for polypharmacy safety it is crucial to identify DDIs and explore their underlying mechanisms. The detection of DDI in the wet lab is expensive and time-consuming, due to the need for experimental research over a large volume of drug combinations. Although many computational methods have been developed to predict DDIs, most of these are incapable of predicting potential DDIs between drugs within the DDI network and new drugs from outside the DDI network. In addition, they are not designed to explore the underlying mechanisms of DDIs and lack interpretative capacity. Thus, here we propose a novel method of GNN-DDI to predict potential DDIs by constructing a five-layer graph attention network to identify k-hops low-dimensional feature representations for each drug from its chemical molecular graph, concatenating all identified features of each drug pair, and inputting them into a MLP predictor to obtain the final DDI prediction score. The experimental results demonstrate that our GNN-DDI is suitable for each of two DDI predicting scenarios, namely the potential DDIs among known drugs in the DDI network and those between drugs within the DDI network and new drugs from outside DDI network. The case study indicates that our method can explore the specific drug substructures that lead to the potential DDIs, which helps to improve interpretability and discover the underlying interaction mechanisms of drug pairs.
Collapse
|
12
|
Firoozbakht F, Yousefi B, Schwikowski B. An overview of machine learning methods for monotherapy drug response prediction. Brief Bioinform 2022; 23:bbab408. [PMID: 34619752 PMCID: PMC8769705 DOI: 10.1093/bib/bbab408] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/25/2021] [Accepted: 09/06/2021] [Indexed: 12/11/2022] Open
Abstract
For an increasing number of preclinical samples, both detailed molecular profiles and their responses to various drugs are becoming available. Efforts to understand, and predict, drug responses in a data-driven manner have led to a proliferation of machine learning (ML) methods, with the longer term ambition of predicting clinical drug responses. Here, we provide a uniquely wide and deep systematic review of the rapidly evolving literature on monotherapy drug response prediction, with a systematic characterization and classification that comprises more than 70 ML methods in 13 subclasses, their input and output data types, modes of evaluation, and code and software availability. ML experts are provided with a fundamental understanding of the biological problem, and how ML methods are configured for it. Biologists and biomedical researchers are introduced to the basic principles of applicable ML methods, and their application to the problem of drug response prediction. We also provide systematic overviews of commonly used data sources used for training and evaluation methods.
Collapse
Affiliation(s)
- Farzaneh Firoozbakht
- Systems Biology Group, Department of Computational Biology, Institut Pasteur, Paris, France
| | - Behnam Yousefi
- Systems Biology Group, Department of Computational Biology, Institut Pasteur, Paris, France
- Sorbonne Université, École Doctorale Complexite du Vivant, Paris, France
| | - Benno Schwikowski
- Systems Biology Group, Department of Computational Biology, Institut Pasteur, Paris, France
| |
Collapse
|
13
|
Fu H, Huang F, Liu X, Qiu Y, Zhang W. MVGCN: data integration through multi-view graph convolutional network for predicting links in biomedical bipartite networks. Bioinformatics 2022; 38:426-434. [PMID: 34499148 DOI: 10.1093/bioinformatics/btab651] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 08/07/2021] [Accepted: 09/06/2021] [Indexed: 02/05/2023] Open
Abstract
MOTIVATION There are various interaction/association bipartite networks in biomolecular systems. Identifying unobserved links in biomedical bipartite networks helps to understand the underlying molecular mechanisms of human complex diseases and thus benefits the diagnosis and treatment of diseases. Although a great number of computational methods have been proposed to predict links in biomedical bipartite networks, most of them heavily depend on features and structures involving the bioentities in one specific bipartite network, which limits the generalization capacity of applying the models to other bipartite networks. Meanwhile, bioentities usually have multiple features, and how to leverage them has also been challenging. RESULTS In this study, we propose a novel multi-view graph convolution network (MVGCN) framework for link prediction in biomedical bipartite networks. We first construct a multi-view heterogeneous network (MVHN) by combining the similarity networks with the biomedical bipartite network, and then perform a self-supervised learning strategy on the bipartite network to obtain node attributes as initial embeddings. Further, a neighborhood information aggregation (NIA) layer is designed for iteratively updating the embeddings of nodes by aggregating information from inter- and intra-domain neighbors in every view of the MVHN. Next, we combine embeddings of multiple NIA layers in each view, and integrate multiple views to obtain the final node embeddings, which are then fed into a discriminator to predict the existence of links. Extensive experiments show MVGCN performs better than or on par with baseline methods and has the generalization capacity on six benchmark datasets involving three typical tasks. AVAILABILITY AND IMPLEMENTATION Source code and data can be downloaded from https://github.com/fuhaitao95/MVGCN. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Haitao Fu
- College of Informatics, Huazhong Agricultural University, Wuhan 430070, China
| | - Feng Huang
- College of Informatics, Huazhong Agricultural University, Wuhan 430070, China
| | - Xuan Liu
- College of Informatics, Huazhong Agricultural University, Wuhan 430070, China
| | - Yang Qiu
- College of Informatics, Huazhong Agricultural University, Wuhan 430070, China
| | - Wen Zhang
- College of Informatics, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
14
|
Liu X, Song C, Huang F, Fu H, Xiao W, Zhang W. GraphCDR: a graph neural network method with contrastive learning for cancer drug response prediction. Brief Bioinform 2021; 23:6415314. [PMID: 34727569 DOI: 10.1093/bib/bbab457] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 09/25/2021] [Accepted: 10/07/2021] [Indexed: 12/29/2022] Open
Abstract
Predicting the response of a cancer cell line to a therapeutic drug is an important topic in modern oncology that can help personalized treatment for cancers. Although numerous machine learning methods have been developed for cancer drug response (CDR) prediction, integrating diverse information about cancer cell lines, drugs and their known responses still remains a great challenge. In this paper, we propose a graph neural network method with contrastive learning for CDR prediction. GraphCDR constructs a graph neural network based on multi-omics profiles of cancer cell lines, the chemical structure of drugs and known cancer cell line-drug responses for CDR prediction, while a contrastive learning task is presented as a regularizer within a multi-task learning paradigm to enhance the generalization ability. In the computational experiments, GraphCDR outperforms state-of-the-art methods under different experimental configurations, and the ablation study reveals the key components of GraphCDR: biological features, known cancer cell line-drug responses and contrastive learning are important for the high-accuracy CDR prediction. The experimental analyses imply the predictive power of GraphCDR and its potential value in guiding anti-cancer drug selection.
Collapse
Affiliation(s)
- Xuan Liu
- College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Congzhi Song
- College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Feng Huang
- College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Haitao Fu
- College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wenjie Xiao
- Information School, University of Washington, Washington, 98105, USA
| | - Wen Zhang
- College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| |
Collapse
|
15
|
Yi HC, You ZH, Guo ZH, Huang DS, Chan KCC. Learning Representation of Molecules in Association Network for Predicting Intermolecular Associations. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:2546-2554. [PMID: 32070992 DOI: 10.1109/tcbb.2020.2973091] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
A key aim of post-genomic biomedical research is to systematically understand molecules and their interactions in human cells. Multiple biomolecules coordinate to sustain life activities, and interactions between various biomolecules are interconnected. However, existing studies usually only focusing on associations between two or very limited types of molecules. In this study, we propose a network representation learning based computational framework MAN-SDNE to predict any intermolecular associations. More specifically, we constructed a large-scale molecular association network of multiple biomolecules in human by integrating associations among long non-coding RNA, microRNA, protein, drug, and disease, containing 6,528 molecular nodes, 9 kind of,105,546 associations. And then, the feature of each node is represented by its network proximity and attribute features. Furthermore, these features are used to train Random Forest classifier to predict intermolecular associations. MAN-SDNE achieves a remarkable performance with an AUC of 0.9552 and an AUPR of 0.9338 under five-fold cross-validation. To indicate the ability to predict specific types of interactions, a case study for predicting lncRNA-protein interactions using MAN-SDNE is also executed. Experimental results demonstrate this work offers a systematic insight for understanding the synergistic associations between molecules and complex diseases and provides a network-based computational tool to systematically explore intermolecular interactions.
Collapse
|
16
|
AlQuraishi M, Sorger PK. Differentiable biology: using deep learning for biophysics-based and data-driven modeling of molecular mechanisms. Nat Methods 2021; 18:1169-1180. [PMID: 34608321 PMCID: PMC8793939 DOI: 10.1038/s41592-021-01283-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 08/27/2021] [Indexed: 02/08/2023]
Abstract
Deep learning using neural networks relies on a class of machine-learnable models constructed using 'differentiable programs'. These programs can combine mathematical equations specific to a particular domain of natural science with general-purpose, machine-learnable components trained on experimental data. Such programs are having a growing impact on molecular and cellular biology. In this Perspective, we describe an emerging 'differentiable biology' in which phenomena ranging from the small and specific (for example, one experimental assay) to the broad and complex (for example, protein folding) can be modeled effectively and efficiently, often by exploiting knowledge about basic natural phenomena to overcome the limitations of sparse, incomplete and noisy data. By distilling differentiable biology into a small set of conceptual primitives and illustrative vignettes, we show how it can help to address long-standing challenges in integrating multimodal data from diverse experiments across biological scales. This promises to benefit fields as diverse as biophysics and functional genomics.
Collapse
Affiliation(s)
- Mohammed AlQuraishi
- Department of Systems Biology, Columbia University, New York, NY, USA.
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
17
|
Rydzewski NR, Peterson E, Lang JM, Yu M, Laura Chang S, Sjöström M, Bakhtiar H, Song G, Helzer KT, Bootsma ML, Chen WS, Shrestha RM, Zhang M, Quigley DA, Aggarwal R, Small EJ, Wahl DR, Feng FY, Zhao SG. Predicting cancer drug TARGETS - TreAtment Response Generalized Elastic-neT Signatures. NPJ Genom Med 2021; 6:76. [PMID: 34548481 PMCID: PMC8455625 DOI: 10.1038/s41525-021-00239-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/23/2021] [Indexed: 12/14/2022] Open
Abstract
We are now in an era of molecular medicine, where specific DNA alterations can be used to identify patients who will respond to specific drugs. However, there are only a handful of clinically used predictive biomarkers in oncology. Herein, we describe an approach utilizing in vitro DNA and RNA sequencing and drug response data to create TreAtment Response Generalized Elastic-neT Signatures (TARGETS). We trained TARGETS drug response models using Elastic-Net regression in the publicly available Genomics of Drug Sensitivity in Cancer (GDSC) database. Models were then validated on additional in-vitro data from the Cancer Cell Line Encyclopedia (CCLE), and on clinical samples from The Cancer Genome Atlas (TCGA) and Stand Up to Cancer/Prostate Cancer Foundation West Coast Prostate Cancer Dream Team (WCDT). First, we demonstrated that all TARGETS models successfully predicted treatment response in the separate in-vitro CCLE treatment response dataset. Next, we evaluated all FDA-approved biomarker-based cancer drug indications in TCGA and demonstrated that TARGETS predictions were concordant with established clinical indications. Finally, we performed independent clinical validation in the WCDT and found that the TARGETS AR signaling inhibitors (ARSI) signature successfully predicted clinical treatment response in metastatic castration-resistant prostate cancer with a statistically significant interaction between the TARGETS score and PSA response (p = 0.0252). TARGETS represents a pan-cancer, platform-independent approach to predict response to oncologic therapies and could be used as a tool to better select patients for existing therapies as well as identify new indications for testing in prospective clinical trials.
Collapse
Affiliation(s)
| | - Erik Peterson
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Joshua M Lang
- Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
- Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Menggang Yu
- Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI, USA
| | - S Laura Chang
- Department of Radiation Oncology, UCSF, San Francisco, CA, USA
| | - Martin Sjöström
- Department of Radiation Oncology, UCSF, San Francisco, CA, USA
| | - Hamza Bakhtiar
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | - Gefei Song
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | - Kyle T Helzer
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | - Matthew L Bootsma
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | - William S Chen
- Department of Radiation Oncology, UCSF, San Francisco, CA, USA
| | | | - Meng Zhang
- Department of Radiation Oncology, UCSF, San Francisco, CA, USA
| | - David A Quigley
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, UCSF, San Francisco, CA, USA
| | - Rahul Aggarwal
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, USA
- Division of Hematology and Oncology, Department of Medicine, UCSF, San Francisco, CA, USA
| | - Eric J Small
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, USA
- Division of Hematology and Oncology, Department of Medicine, UCSF, San Francisco, CA, USA
| | - Daniel R Wahl
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Felix Y Feng
- Department of Radiation Oncology, UCSF, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, USA
- Division of Hematology and Oncology, Department of Medicine, UCSF, San Francisco, CA, USA
- Department of Urology, UCSF, San Francisco, CA, USA
| | - Shuang G Zhao
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA.
- Carbone Cancer Center, University of Wisconsin, Madison, WI, USA.
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
| |
Collapse
|
18
|
Wang X, Yang Y, Li K, Li W, Li F, Peng S. BioERP: biomedical heterogeneous network-based self-supervised representation learning approach for entity relationship predictions. Bioinformatics 2021; 37:4793-4800. [PMID: 34329382 DOI: 10.1093/bioinformatics/btab565] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 07/18/2021] [Accepted: 07/29/2021] [Indexed: 11/14/2022] Open
Abstract
MOTIVATION Predicting entity relationship can greatly benefit important biomedical problems. Recently, a large amount of biomedical heterogeneous networks (BioHNs) are generated and offer opportunities for developing network-based learning approaches to predict relationships among entities. However, current researches slightly explored BioHNs-based self-supervised representation learning methods, and are hard to simultaneously capturing local- and global-level association information among entities. RESULTS In this study, we propose a biomedical heterogeneous network-based self-supervised representation learning approach for entity relationship predictions, termed BioERP. A self-supervised meta path detection mechanism is proposed to train a deep Transformer encoder model that can capture the global structure and semantic feature in BioHNs. Meanwhile, a biomedical entity mask learning strategy is designed to reflect local associations of vertices. Finally, the representations from different task models are concatenated to generate two-level representation vectors for predicting relationships among entities. The results on eight datasets show BioERP outperforms 30 state-of-the-art methods. In particular, BioERP reveals great performance with results close to 1 in terms of AUC and AUPR on the drug-target interaction predictions. In summary, BioERP is a promising bio-entity relationship prediction approach. AVAILABILITY Source code and data can be downloaded from https://github.com/pengsl-lab/BioERP.git. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Xiaoqi Wang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Yaning Yang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Kenli Li
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Wentao Li
- School of Computer Science, National University of Defense Technology, Changsha, 410073, China
| | - Fei Li
- Computer Network Information Center, Chinese Academy of Sciences, Beijing 100850, China
| | - Shaoliang Peng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China.,School of Computer Science, National University of Defense Technology, Changsha, 410073, China.,Peng Cheng Lab, Shenzhen 518000, China
| |
Collapse
|
19
|
Meybodi FY, Eslahchi C. Predicting Anti-Cancer Drug Response by Finding Optimal Subset of Drugs. Bioinformatics 2021; 37:4509-4516. [PMID: 34170297 DOI: 10.1093/bioinformatics/btab466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/26/2021] [Accepted: 06/22/2021] [Indexed: 11/14/2022] Open
Abstract
MOTIVATION One of the most difficult challenges in precision medicine is determining the best treatment strategy for each patient based on personal information. Since drug response prediction in vitro is extremely expensive, time-consuming, and virtually impossible, and because there are so many cell lines and drug data, computational methods are needed. RESULTS MinDrug is a method for predicting anti-cancer drug response which try to identify the best subset of drugs that are the most similar to other drugs. MinDrug predicts the anti-cancer drug response on a new cell line using information from drugs in this subset and their connections to other drugs. MinDrug employs a heuristic star algorithm to identify an optimal subset of drugs and a regression technique known as Elastic-Net approaches to predict anti-cancer drug response in a new cell line. To test MinDrug, we use both statistical and biological methods to assess the selected drugs. MinDrug is also compared to four state-of-the-art approaches using various k-fold cross-validations on two large public datasets: GDSC and CCLE. MinDrug outperforms the other approaches in terms of precision, robustness, and speed. Furthermore, we compare the evaluation results of all the approaches with an external dataset with a statistical distribution that is not exactly the same as the training data. The results show that MinDrug continues to outperform the other approaches. AVAILABILITY MinDrug's source code can be found at https://github.com/yassaee/MinDrug. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Fatemeh Yassaee Meybodi
- Department of Computer and Data Sciences, Faculty of Mathematical Sciences, Shahid Beheshti University, Tehran, Iran
| | - Changiz Eslahchi
- Department of Computer and Data Sciences, Faculty of Mathematical Sciences, Shahid Beheshti University, Tehran, Iran.,School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| |
Collapse
|
20
|
Huang S, Hu P, Lakowski TM. Predicting breast cancer drug response using a multiple-layer cell line drug response network model. BMC Cancer 2021; 21:648. [PMID: 34059012 PMCID: PMC8166022 DOI: 10.1186/s12885-021-08359-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 05/13/2021] [Indexed: 01/04/2023] Open
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. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08359-6.
Collapse
Affiliation(s)
- Shujun Huang
- College of Pharmacy, University of Manitoba, Apotex Centre, 750 McDermot Avenue, Winnipeg, Manitoba, R3E 0T5, Canada
| | - Pingzhao Hu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Room 308 - Basic Medical Sciences Building, 745 Bannatyne Avenue, Winnipeg, Manitoba, R3E 0J9, Canada. .,Cancer Care Manitoba Research Institute, 675 McDermot Avenue, Winnipeg, Manitoba, R3E 0V9, Canada.
| | - Ted M Lakowski
- College of Pharmacy, University of Manitoba, Apotex Centre, 750 McDermot Avenue, Winnipeg, Manitoba, R3E 0T5, Canada.
| |
Collapse
|
21
|
Tan X, Yu Y, Duan K, Zhang J, Sun P, Sun H. Current Advances and Limitations of Deep Learning in Anticancer Drug Sensitivity Prediction. Curr Top Med Chem 2021; 20:1858-1867. [PMID: 32648840 DOI: 10.2174/1568026620666200710101307] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/02/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023]
Abstract
Anticancer drug screening can accelerate drug discovery to save the lives of cancer patients, but cancer heterogeneity makes this screening challenging. The prediction of anticancer drug sensitivity is useful for anticancer drug development and the identification of biomarkers of drug sensitivity. Deep learning, as a branch of machine learning, is an important aspect of in silico research. Its outstanding computational performance means that it has been used for many biomedical purposes, such as medical image interpretation, biological sequence analysis, and drug discovery. Several studies have predicted anticancer drug sensitivity based on deep learning algorithms. The field of deep learning has made progress regarding model performance and multi-omics data integration. However, deep learning is limited by the number of studies performed and data sources available, so it is not perfect as a pre-clinical approach for use in the anticancer drug screening process. Improving the performance of deep learning models is a pressing issue for researchers. In this review, we introduce the research of anticancer drug sensitivity prediction and the use of deep learning in this research area. To provide a reference for future research, we also review some common data sources and machine learning methods. Lastly, we discuss the advantages and disadvantages of deep learning, as well as the limitations and future perspectives regarding this approach.
Collapse
Affiliation(s)
- Xian Tan
- School of Information Science and Technology, Northeast Normal University, Changchun 130117, China
| | - Yang Yu
- School of Information Science and Technology, Northeast Normal University, Changchun 130117, China
| | - Kaiwen Duan
- School of Information Science and Technology, Northeast Normal University, Changchun 130117, China
| | - Jingbo Zhang
- School of Information Science and Technology, Northeast Normal University, Changchun 130117, China
| | - Pingping Sun
- School of Information Science and Technology, Northeast Normal University, Changchun 130117, China
| | - Hui Sun
- College of Humanities and Sciences of Northeast Normal University, Changchun 130117, China
| |
Collapse
|
22
|
Ahmadi Moughari F, Eslahchi C. A computational method for drug sensitivity prediction of cancer cell lines based on various molecular information. PLoS One 2021; 16:e0250620. [PMID: 33914775 PMCID: PMC8084246 DOI: 10.1371/journal.pone.0250620] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 04/09/2021] [Indexed: 12/29/2022] Open
Abstract
Determining sensitive drugs for a patient is one of the most critical problems in precision medicine. Using genomic profiles of the tumor and drug information can help in tailoring the most efficient treatment for a patient. In this paper, we proposed a classification machine learning approach that predicts the sensitive/resistant drugs for a cell line. It can be performed by using both drug and cell line similarities, one of the cell line or drug similarities, or even not using any similarity information. This paper investigates the influence of using previously defined as well as two newly introduced similarities on predicting anti-cancer drug sensitivity. The proposed method uses max concentration thresholds for assigning drug responses to class labels. Its performance was evaluated using stratified five-fold cross-validation on cell line-drug pairs in two datasets. Assessing the predictive powers of the proposed model and three sets of methods, including state-of-the-art classification methods, state-of-the-art regression methods, and off-the-shelf classification machine learning approaches shows that the proposed method outperforms other methods. Moreover, The efficiency of the model is evaluated in tissue-specific conditions. Besides, the novel sensitive associations predicted by this model were verified by several supportive evidence in the literature and reliable database. Therefore, the proposed model can efficiently be used in predicting anti-cancer drug sensitivity. Material and implementation are available at https://github.com/fahmadimoughari/CDSML.
Collapse
Affiliation(s)
- Fatemeh Ahmadi Moughari
- Department of Computer and Data Sciences, Faculty of Mathematical Sciences, Shahid Beheshti University, Tehran, Iran
| | - Changiz Eslahchi
- Department of Computer and Data Sciences, Faculty of Mathematical Sciences, Shahid Beheshti University, Tehran, Iran
- School of Biological Sciences, Institute for Research in Fundamental Sciences(IPM), Tehran, Iran
| |
Collapse
|
23
|
Ahmed KT, Park S, Jiang Q, Yeu Y, Hwang T, Zhang W. Network-based drug sensitivity prediction. BMC Med Genomics 2020; 13:193. [PMID: 33371891 PMCID: PMC7771088 DOI: 10.1186/s12920-020-00829-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022] Open
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.
Collapse
Affiliation(s)
- Khandakar Tanvir Ahmed
- Department of Computer Science, University of Central Florida, 4000 Central Florida Blvd, Orlando, FL, 32816, USA
| | - Sunho Park
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, 9211 Euclid Ave, Cleveland, OH, 44106, USA
| | - Qibing Jiang
- Department of Computer Science, University of Central Florida, 4000 Central Florida Blvd, Orlando, FL, 32816, USA
| | - Yunku Yeu
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, 9211 Euclid Ave, Cleveland, OH, 44106, USA
| | - TaeHyun Hwang
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, 9211 Euclid Ave, Cleveland, OH, 44106, USA
| | - Wei Zhang
- Department of Computer Science, University of Central Florida, 4000 Central Florida Blvd, Orlando, FL, 32816, USA.
| |
Collapse
|
24
|
Huang LC, Yeung W, Wang Y, Cheng H, Venkat A, Li S, Ma P, Rasheed K, Kannan N. Quantitative Structure-Mutation-Activity Relationship Tests (QSMART) model for protein kinase inhibitor response prediction. BMC Bioinformatics 2020; 21:520. [PMID: 33183223 PMCID: PMC7664030 DOI: 10.1186/s12859-020-03842-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 10/27/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Protein kinases are a large family of druggable proteins that are genomically and proteomically altered in many human cancers. Kinase-targeted drugs are emerging as promising avenues for personalized medicine because of the differential response shown by altered kinases to drug treatment in patients and cell-based assays. However, an incomplete understanding of the relationships connecting genome, proteome and drug sensitivity profiles present a major bottleneck in targeting kinases for personalized medicine. RESULTS In this study, we propose a multi-component Quantitative Structure-Mutation-Activity Relationship Tests (QSMART) model and neural networks framework for providing explainable models of protein kinase inhibition and drug response ([Formula: see text]) profiles in cell lines. Using non-small cell lung cancer as a case study, we show that interaction terms that capture associations between drugs, pathways, and mutant kinases quantitatively contribute to the response of two EGFR inhibitors (afatinib and lapatinib). In particular, protein-protein interactions associated with the JNK apoptotic pathway, associations between lung development and axon extension, and interaction terms connecting drug substructures and the volume/charge of mutant residues at specific structural locations contribute significantly to the observed [Formula: see text] values in cell-based assays. CONCLUSIONS By integrating multi-omics data in the QSMART model, we not only predict drug responses in cancer cell lines with high accuracy but also identify features and explainable interaction terms contributing to the accuracy. Although we have tested our multi-component explainable framework on protein kinase inhibitors, it can be extended across the proteome to investigate the complex relationships connecting genotypes and drug sensitivity profiles.
Collapse
Affiliation(s)
- Liang-Chin Huang
- Institute of Bioinformatics, University of Georgia, 120 Green St., Athens, GA 30602 USA
| | - Wayland Yeung
- Institute of Bioinformatics, University of Georgia, 120 Green St., Athens, GA 30602 USA
| | - Ye Wang
- Department of Statistics, University of Georgia, 310 Herty Drive, Athens, GA 30602 USA
| | - Huimin Cheng
- Department of Statistics, University of Georgia, 310 Herty Drive, Athens, GA 30602 USA
| | - Aarya Venkat
- Department of Biochemistry and Molecular Biology, 120 Green St., Athens, GA 30602 USA
| | - Sheng Li
- Department of Computer Science, 415 Boyd Graduate Studies Research Center, Athens, GA 30602 USA
| | - Ping Ma
- Department of Statistics, University of Georgia, 310 Herty Drive, Athens, GA 30602 USA
| | - Khaled Rasheed
- Department of Computer Science, 415 Boyd Graduate Studies Research Center, Athens, GA 30602 USA
| | - Natarajan Kannan
- Institute of Bioinformatics, University of Georgia, 120 Green St., Athens, GA 30602 USA
- Department of Biochemistry and Molecular Biology, 120 Green St., Athens, GA 30602 USA
| |
Collapse
|
25
|
Shen C, Luo J, Ouyang W, Ding P, Wu H. Identification of Small Molecule–miRNA Associations with Graph Regularization Techniques in Heterogeneous Networks. J Chem Inf Model 2020; 60:6709-6721. [DOI: 10.1021/acs.jcim.0c00975] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Cong Shen
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410083, China
| | - Jiawei Luo
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410083, China
| | - Wenjue Ouyang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410083, China
| | - Pingjian Ding
- School of Computer Science, University of South China, Hengyang 421001, China
| | - Hao Wu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410083, China
| |
Collapse
|
26
|
Clayton EA, Pujol TA, McDonald JF, Qiu P. Leveraging TCGA gene expression data to build predictive models for cancer drug response. BMC Bioinformatics 2020; 21:364. [PMID: 32998700 PMCID: PMC7526215 DOI: 10.1186/s12859-020-03690-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Machine learning has been utilized to predict cancer drug response from multi-omics data generated from sensitivities of cancer cell lines to different therapeutic compounds. Here, we build machine learning models using gene expression data from patients' primary tumor tissues to predict whether a patient will respond positively or negatively to two chemotherapeutics: 5-Fluorouracil and Gemcitabine. RESULTS We focused on 5-Fluorouracil and Gemcitabine because based on our exclusion criteria, they provide the largest numbers of patients within TCGA. Normalized gene expression data were clustered and used as the input features for the study. We used matching clinical trial data to ascertain the response of these patients via multiple classification methods. Multiple clustering and classification methods were compared for prediction accuracy of drug response. Clara and random forest were found to be the best clustering and classification methods, respectively. The results show our models predict with up to 86% accuracy; despite the study's limitation of sample size. We also found the genes most informative for predicting drug response were enriched in well-known cancer signaling pathways and highlighted their potential significance in chemotherapy prognosis. CONCLUSIONS Primary tumor gene expression is a good predictor of cancer drug response. Investment in larger datasets containing both patient gene expression and drug response is needed to support future work of machine learning models. Ultimately, such predictive models may aid oncologists with making critical treatment decisions.
Collapse
Affiliation(s)
- Evan A. Clayton
- Integrated Cancer Research Center, School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA USA
| | - Toyya A. Pujol
- School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA USA
| | - John F. McDonald
- Integrated Cancer Research Center, School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA USA
| | - Peng Qiu
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 950 Atlantic Dr NW, 30332-0230, Atlanta, GA 404-385-1656 USA
| |
Collapse
|
27
|
Xi J, Yuan X, Wang M, Li A, Li X, Huang Q. Inferring subgroup-specific driver genes from heterogeneous cancer samples via subspace learning with subgroup indication. Bioinformatics 2020; 36:1855-1863. [PMID: 31626284 DOI: 10.1093/bioinformatics/btz793] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/23/2019] [Accepted: 10/16/2019] [Indexed: 11/14/2022] Open
Abstract
MOTIVATION Detecting driver genes from gene mutation data is a fundamental task for tumorigenesis research. Due to the fact that cancer is a heterogeneous disease with various subgroups, subgroup-specific driver genes are the key factors in the development of precision medicine for heterogeneous cancer. However, the existing driver gene detection methods are not designed to identify subgroup specificities of their detected driver genes, and therefore cannot indicate which group of patients is associated with the detected driver genes, which is difficult to provide specifically clinical guidance for individual patients. RESULTS By incorporating the subspace learning framework, we propose a novel bioinformatics method called DriverSub, which can efficiently predict subgroup-specific driver genes in the situation where the subgroup annotations are not available. When evaluated by simulation datasets with known ground truth and compared with existing methods, DriverSub yields the best prediction of driver genes and the inference of their related subgroups. When we apply DriverSub on the mutation data of real heterogeneous cancers, we can observe that the predicted results of DriverSub are highly enriched for experimentally validated known driver genes. Moreover, the subgroups inferred by DriverSub are significantly associated with the annotated molecular subgroups, indicating its capability of predicting subgroup-specific driver genes. AVAILABILITY AND IMPLEMENTATION The source code is publicly available at https://github.com/JianingXi/DriverSub. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Jianing Xi
- School of Mechanical Engineering , Northwestern Polytechnical University, Xi'an, 710072, China.,Center for OPTical IMagery Analysis and Learning (OPTIMAL), Northwestern Polytechnical University, Xi'an, 710072, China
| | - Xiguo Yuan
- School of Computer Science and Technology, Xidian University, Xi'an 710071, China
| | - Minghui Wang
- School of Information Science and Technology, University of Science and Technology of China, Hefei 230027, China
| | - Ao Li
- School of Information Science and Technology, University of Science and Technology of China, Hefei 230027, China
| | - Xuelong Li
- Center for OPTical IMagery Analysis and Learning (OPTIMAL), Northwestern Polytechnical University, Xi'an, 710072, China.,School of Computer Science, Northwestern Polytechnical University, Xi'an 710072, China
| | - Qinghua Huang
- School of Mechanical Engineering , Northwestern Polytechnical University, Xi'an, 710072, China.,Center for OPTical IMagery Analysis and Learning (OPTIMAL), Northwestern Polytechnical University, Xi'an, 710072, China
| |
Collapse
|
28
|
Huang J, Chen J, Zhang B, Zhu L, Cai H. Evaluation of gene-drug common module identification methods using pharmacogenomics data. Brief Bioinform 2020; 22:5860683. [PMID: 32591780 DOI: 10.1093/bib/bbaa087] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/06/2020] [Accepted: 04/23/2020] [Indexed: 01/21/2023] Open
Abstract
Accurately identifying the interactions between genomic factors and the response of cancer drugs plays important roles in drug discovery, drug repositioning and cancer treatment. A number of studies revealed that interactions between genes and drugs were 'many-genes-to-many drugs' interactions, i.e. common modules, opposed to 'one-gene-to-one-drug' interactions. Such modules fully explain the interactions between complex biological regulatory mechanisms and cancer drugs. However, strategies for effectively and robustly identifying the underlying common modules among pharmacogenomics data remain to be improved. In this paper, we aim to provide a detailed evaluation of three categories of state-of-the-art common module identification techniques from a machine learning perspective, including non-negative matrix factorization (NMF), partial least squares (PLS) and network analyses. We first evaluate the performance of six methods, namely SNMNMF, NetNMF, SNPLS, O2PLS, NSBM and HOGMMNC, using two series of simulated data sets with different noise levels and outlier ratios. Then, we conduct experiments using a real world data set of 2091 genes and 101 drugs in 392 cancer cell lines and compare the real experimental results from the aspect of biological process term enrichment, gene-drug and drug-drug interactions. Finally, we present interesting findings from our evaluation study and discuss the advantages and drawbacks of each method. Supplementary information: Supplementary file is available at Briefings in Bioinformatics online.
Collapse
Affiliation(s)
- Jie Huang
- South China University of Technology, School of Computer Science and Engineering, Guangzhou, 510006, China
| | - Jiazhou Chen
- South China University of Technology, School of Computer Science and Engineering, Guangzhou, 510006, China
| | - Bin Zhang
- South China University of Technology, School of Computer Science and Engineering, Guangzhou, 510006, China
| | - Lei Zhu
- South China University of Technology, School of Computer Science and Engineering, Guangzhou, 510006, China
| | - Hongmin Cai
- South China University of Technology, School of Computer Science and Engineering, Guangzhou, 510006, China
| |
Collapse
|
29
|
Zheng H, Zhang G, Zhang L, Wang Q, Li H, Han Y, Xie L, Yan Z, Li Y, An Y, Dong H, Zhu W, Guo X. Comprehensive Review of Web Servers and Bioinformatics Tools for Cancer Prognosis Analysis. Front Oncol 2020; 10:68. [PMID: 32117725 PMCID: PMC7013087 DOI: 10.3389/fonc.2020.00068] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/15/2020] [Indexed: 01/10/2023] Open
Abstract
Prognostic biomarkers are of great significance to predict the outcome of patients with cancer, to guide the clinical treatments, to elucidate tumorigenesis mechanisms, and offer the opportunity of identifying therapeutic targets. To screen and develop prognostic biomarkers, high throughput profiling methods including gene microarray and next-generation sequencing have been widely applied and shown great success. However, due to the lack of independent validation, only very few prognostic biomarkers have been applied for clinical practice. In order to cross-validate the reliability of potential prognostic biomarkers, some groups have collected the omics datasets (i.e., epigenetics/transcriptome/proteome) with relative follow-up data (such as OS/DSS/PFS) of clinical samples from different cohorts, and developed the easy-to-use online bioinformatics tools and web servers to assist the biomarker screening and validation. These tools and web servers provide great convenience for the development of prognostic biomarkers, for the study of molecular mechanisms of tumorigenesis and progression, and even for the discovery of important therapeutic targets. Aim to help researchers to get a quick learning and understand the function of these tools, the current review delves into the introduction of the usage, characteristics and algorithms of tools, and web servers, such as LOGpc, KM plotter, GEPIA, TCPA, OncoLnc, PrognoScan, MethSurv, SurvExpress, UALCAN, etc., and further help researchers to select more suitable tools for their own research. In addition, all the tools introduced in this review can be reached at http://bioinfo.henu.edu.cn/WebServiceList.html.
Collapse
Affiliation(s)
- Hong Zheng
- Cell Signal Transduction Laboratory, Bioinformatics Center, School of Basic Medical Sciences, School of Software, Institute of Biomedical Informatics, Henan University, Kaifeng, China
| | - Guosen Zhang
- Cell Signal Transduction Laboratory, Bioinformatics Center, School of Basic Medical Sciences, School of Software, Institute of Biomedical Informatics, Henan University, Kaifeng, China
| | - Lu Zhang
- Cell Signal Transduction Laboratory, Bioinformatics Center, School of Basic Medical Sciences, School of Software, Institute of Biomedical Informatics, Henan University, Kaifeng, China
| | - Qiang Wang
- Cell Signal Transduction Laboratory, Bioinformatics Center, School of Basic Medical Sciences, School of Software, Institute of Biomedical Informatics, Henan University, Kaifeng, China
| | - Huimin Li
- Cell Signal Transduction Laboratory, Bioinformatics Center, School of Basic Medical Sciences, School of Software, Institute of Biomedical Informatics, Henan University, Kaifeng, China
| | - Yali Han
- Cell Signal Transduction Laboratory, Bioinformatics Center, School of Basic Medical Sciences, School of Software, Institute of Biomedical Informatics, Henan University, Kaifeng, China
| | - Longxiang Xie
- Cell Signal Transduction Laboratory, Bioinformatics Center, School of Basic Medical Sciences, School of Software, Institute of Biomedical Informatics, Henan University, Kaifeng, China
| | - Zhongyi Yan
- Cell Signal Transduction Laboratory, Bioinformatics Center, School of Basic Medical Sciences, School of Software, Institute of Biomedical Informatics, Henan University, Kaifeng, China
| | - Yongqiang Li
- Cell Signal Transduction Laboratory, Bioinformatics Center, School of Basic Medical Sciences, School of Software, Institute of Biomedical Informatics, Henan University, Kaifeng, China
| | - Yang An
- Cell Signal Transduction Laboratory, Bioinformatics Center, School of Basic Medical Sciences, School of Software, Institute of Biomedical Informatics, Henan University, Kaifeng, China
| | - Huan Dong
- Cell Signal Transduction Laboratory, Bioinformatics Center, School of Basic Medical Sciences, School of Software, Institute of Biomedical Informatics, Henan University, Kaifeng, China
| | - Wan Zhu
- Department of Anesthesia, Stanford University, Stanford, CA, United States
| | - Xiangqian Guo
- Cell Signal Transduction Laboratory, Bioinformatics Center, School of Basic Medical Sciences, School of Software, Institute of Biomedical Informatics, Henan University, Kaifeng, China
| |
Collapse
|
30
|
Liu Q, Muglia LJ, Huang LF. Network as a Biomarker: A Novel Network-Based Sparse Bayesian Machine for Pathway-Driven Drug Response Prediction. Genes (Basel) 2019; 10:genes10080602. [PMID: 31405013 PMCID: PMC6723660 DOI: 10.3390/genes10080602] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 01/17/2023] Open
Abstract
With the advances in different biological networks including gene regulation, gene co-expression, protein–protein interaction networks, and advanced approaches for network reconstruction, analysis, and interpretation, it is possible to discover reliable and accurate molecular network-based biomarkers for monitoring cancer treatment. Such efforts will also pave the way toward the realization of biomarker-driven personalized medicine against cancer. Previously, we have reconstructed disease-specific driver signaling networks using multi-omics profiles and cancer signaling pathway data. In this study, we developed a network-based sparse Bayesian machine (NBSBM) approach, using previously derived disease-specific driver signaling networks to predict cancer cell responses to drugs. NBSBM made use of the information encoded in a disease-specific (differentially expressed) network to improve its prediction performance in problems with a reduced amount of training data and a very high-dimensional feature space. Sparsity in NBSBM is favored by a spike and slab prior distribution, which is combined with a Markov random field prior that encodes the network of feature dependencies. Gene features that are connected in the network are assumed to be both relevant and irrelevant to drug responses. We compared the proposed method with network-based support vector machine (NBSVM) approaches and found that the NBSBM approach could achieve much better accuracy than the other two NBSVM methods. The gene modules selected from the disease-specific driver networks for predicting drug sensitivity might be directly involved in drug sensitivity or resistance. This work provides a disease-specific network-based drug sensitivity prediction approach and can uncover the potential mechanisms of the action of drugs by selecting the most predictive sub-networks from the disease-specific network.
Collapse
Affiliation(s)
- Qi Liu
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Louis J Muglia
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lei Frank Huang
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA.
- Department of Information Science, School of Mathematical Sciences and LAMA, Peking University, Beijing 100871, China.
| |
Collapse
|
31
|
Xu Y, Dong Q, Li F, Xu Y, Hu C, Wang J, Shang D, Zheng X, Yang H, Zhang C, Shao M, Meng M, Xiong Z, Li X, Zhang Y. Identifying subpathway signatures for individualized anticancer drug response by integrating multi-omics data. J Transl Med 2019; 17:255. [PMID: 31387579 PMCID: PMC6685260 DOI: 10.1186/s12967-019-2010-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 07/31/2019] [Indexed: 12/19/2022] Open
Abstract
Background Individualized drug response prediction is vital for achieving personalized treatment of cancer and moving precision medicine forward. Large-scale multi-omics profiles provide unprecedented opportunities for precision cancer therapy. Methods In this study, we propose a pipeline to identify subpathway signatures for anticancer drug response of individuals by integrating the comprehensive contributions of multiple genetic and epigenetic (gene expression, copy number variation and DNA methylation) alterations. Results Totally, 46 subpathway signatures associated with individual responses to different anticancer drugs were identified based on five cancer-drug response datasets. We have validated the reliability of subpathway signatures in two independent datasets. Furthermore, we also demonstrated these multi-omics subpathway signatures could significantly improve the performance of anticancer drug response prediction. In-depth analysis of these 46 subpathway signatures uncovered the essential roles of three omics types and the functional associations underlying different anticancer drug responses. Patient stratification based on subpathway signatures involved in anticancer drug response identified subtypes with different clinical outcomes, implying their potential roles as prognostic biomarkers. In addition, a landscape of subpathways associated with cellular responses to 191 anticancer drugs from CellMiner was provided and the mechanism similarity of drug action was accurately unclosed based on these subpathways. Finally, we constructed a user-friendly web interface-CancerDAP (http://bio-bigdata.hrbmu.edu.cn/CancerDAP/) available to explore 2751 subpathways relevant with 191 anticancer drugs response. Conclusions Taken together, our study identified and systematically characterized subpathway signatures for individualized anticancer drug response prediction, which may promote the precise treatment of cancer and the study for molecular mechanisms of drug actions. Electronic supplementary material The online version of this article (10.1186/s12967-019-2010-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yanjun Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Qun Dong
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Feng Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yingqi Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Congxue Hu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Jingwen Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Desi Shang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Xuan Zheng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Haixiu Yang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Chunlong Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Mengting Shao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Mohan Meng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Zhiying Xiong
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China.
| | - Yunpeng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
32
|
Milan T, Canaj H, Villeneuve C, Ghosh A, Barabé F, Cellot S, Wilhelm BT. Pediatric leukemia: Moving toward more accurate models. Exp Hematol 2019; 74:1-12. [PMID: 31154068 DOI: 10.1016/j.exphem.2019.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/19/2019] [Accepted: 05/22/2019] [Indexed: 02/07/2023]
Abstract
Leukemia is a complex genetic disease caused by errors in differentiation, growth, and apoptosis of hematopoietic cells in either lymphoid or myeloid lineages. Large-scale genomic characterization of thousands of leukemia patients has produced a tremendous amount of data that have enabled a better understanding of the differences between adult and pediatric patients. For instance, although phenotypically similar, pediatric and adult myeloid leukemia patients differ in their mutational profiles, typically involving either chromosomal translocations or recurrent single-base-pair mutations, respectively. To elucidate the molecular mechanisms underlying the biology of this cancer, continual efforts have been made to develop more contextually and biologically relevant experimental models. Leukemic cell lines, for example, provide an inexpensive and tractable model but often fail to recapitulate critical aspects of tumor biology. Likewise, murine leukemia models of leukemia have been highly informative but also do not entirely reproduce the human disease. More recent advances in the development of patient-derived xenografts (PDXs) or human models of leukemias are poised to provide a more comprehensive, and biologically relevant, approach to directly assess the impact of the in vivo environment on human samples. In this review, the advantages and limitations of the various current models used to functionally define the genetic requirements of leukemogenesis are discussed.
Collapse
MESH Headings
- Adolescent
- Animals
- Cell Differentiation
- Child
- Child, Preschool
- Female
- Heterografts
- Humans
- Infant
- Infant, Newborn
- Leukemia, Myeloid/genetics
- Leukemia, Myeloid/pathology
- Leukemia, Myeloid/therapy
- Male
- Mice
- Neoplasm Transplantation
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/therapy
- Translocation, Genetic
Collapse
Affiliation(s)
- Thomas Milan
- Laboratory for High Throughput Biology, Institute for Research in Immunology and Cancer, Montréal, QC, Canada
| | - Hera Canaj
- Laboratory for High Throughput Biology, Institute for Research in Immunology and Cancer, Montréal, QC, Canada
| | - Chloe Villeneuve
- Laboratory for High Throughput Biology, Institute for Research in Immunology and Cancer, Montréal, QC, Canada
| | - Aditi Ghosh
- Laboratory for High Throughput Biology, Institute for Research in Immunology and Cancer, Montréal, QC, Canada
| | - Frédéric Barabé
- Centre de recherche en infectiologie du CHUL, Centre de recherche du CHU de Québec, Quebec City, QC, Canada; CHU de Québec Hôpital Enfant-Jésus, Quebec City, QC, Canada; Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - Sonia Cellot
- Division of Hematology, Department of Pediatrics, Ste-Justine Hospital, Montréal, Université de Montréal, Montréal, QC, Canada
| | - Brian T Wilhelm
- Laboratory for High Throughput Biology, Institute for Research in Immunology and Cancer, Montréal, QC, Canada; Department of Medicine, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
33
|
Feng X, Wang E, Cui Q. Gene Expression-Based Predictive Markers for Paclitaxel Treatment in ER+ and ER- Breast Cancer. Front Genet 2019; 10:156. [PMID: 30881385 PMCID: PMC6405635 DOI: 10.3389/fgene.2019.00156] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 02/13/2019] [Indexed: 12/29/2022] Open
Abstract
One of the objectives of precision oncology is to identify patient’s responsiveness to a given treatment and prevent potential overtreatments through molecular profiling. Predictive gene expression biomarkers are a promising and practical means to this purpose. The overall response rate of paclitaxel drugs in breast cancer has been reported to be in the range of 20–60% and is in the even lower range for ER-positive patients. Predicting responsiveness of breast cancer patients, either ER-positive or ER-negative, to paclitaxel treatment could prevent individuals with poor response to the therapy from undergoing excess exposure to the agent. In this study, we identified six sets of gene signatures whose gene expression profiles could robustly predict nonresponding patients with precisions more than 94% and recalls more than 93% on various discovery datasets (n = 469 for the largest set) and independent validation datasets (n = 278), using the previously developed Multiple Survival Screening algorithm, a random-sampling-based methodology. The gene signatures reported were stable regardless of half of the discovery datasets being swapped, demonstrating their robustness. We also reported a set of optimizations that enabled the algorithm to train on small-scale computational resources. The gene signatures and optimized methodology described in this study could be used for identifying unresponsiveness in patients of ER-positive or ER-negative breast cancers.
Collapse
Affiliation(s)
- Xiaowen Feng
- Department of Biomedical Informatics, School of Basic Medical Sciences, MOE Key Lab of Cardiovascular Sciences, Peking University, Beijing, China.,Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Edwin Wang
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Qinghua Cui
- Department of Biomedical Informatics, School of Basic Medical Sciences, MOE Key Lab of Cardiovascular Sciences, Peking University, Beijing, China
| |
Collapse
|