1
|
Zhang Z, Qiu J, Zheng J, Yu Z, Su L, Lin Q, Zhang C, Liao K. Enhanced Regioselectivity Prediction of sp 2 C-H Halogenation via Negative Data Augmentation and Multimodel Integration. J Chem Inf Model 2025; 65:3420-3430. [PMID: 40111160 DOI: 10.1021/acs.jcim.5c00281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Efficient molecular editing is pivotal in synthetic chemistry, especially for developing drugs, materials, and high-value chemicals. Electrophilic aromatic substitution (SEAr) reactions, specifically sp2 C-H halogenation, face significant challenges due to electronic and steric factors, necessitating extensive trial-and-error. This study introduces an innovative machine learning-based model to predict halogenation sites in SEAr reactions, achieving an average accuracy of 93% in 5-fold cross-validation. Employing ensemble techniques, particularly AutoGluon-Tabular (AG), the model demonstrates broad applicability across various aromatic halides, enhancing its utility in drug design, materials science, and more. By reducing experimental uncertainty and optimizing synthetic pathways, this model saves considerable time and resources, thereby accelerating innovation in synthetic chemistry.
Collapse
Affiliation(s)
- Zhiting Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou 511436, China
- Guangzhou National Laboratory, Guangzhou 510005, China
- Southern University of Science and Technology, Shenzhen 518055, China
| | - Jia Qiu
- Guangzhou National Laboratory, Guangzhou 510005, China
| | | | - Zhunzhun Yu
- Guangzhou National Laboratory, Guangzhou 510005, China
| | - Lebin Su
- Guangzhou National Laboratory, Guangzhou 510005, China
| | - Qianghua Lin
- Guangzhou National Laboratory, Guangzhou 510005, China
| | | | - Kuangbiao Liao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou 511436, China
- Guangzhou National Laboratory, Guangzhou 510005, China
| |
Collapse
|
2
|
Tran TO, Nguyen TH, Nguyen TT, Le NQK. MLG2Net: Molecular Global Graph Network for Drug Response Prediction in Lung Cancer Cell Lines. J Med Syst 2025; 49:47. [PMID: 40208442 DOI: 10.1007/s10916-025-02182-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Drug response prediction (DRP) is a central task in the era of precision medicine. Over the past decade, the emergence of deep learning (DL) has greatly contributed to addressing DRP challenges. Notably, the prediction of DRP for cancer cell lines benefits significantly from data availability for model development. However, an effective predictive model is still challenging due to issues with data quality, high-dimensional data, and multi-omics data integration. In this study, we introduce MLG2Net, a deep-learning model inspired by graph neural networks designed to predict DRP in lung cancer cell lines based on pharmacogenomics data. Our model comprises two key components: drug SMILES described by local and global graph networks and cell line genomics are illustrated as a map. Our results show that MLG2Net outperforms three reference graph networks. MLG2Net performance reached a Pearson coefficient correlation (C C p ) of 0.8616 and a root mean square error (RMSE) of 2.94e-6 in predicting drug responses for Lung Adenocarcinoma (LUAD) cell lines. Subsequent testing on the Lung Squamous Cell Carcinoma (LUSC) dataset reveals lower performance (C C p : 0.7999, RMSE: 4.08e-6), attributed to the dataset's smaller size influencing model capacity. Moreover, we assessed the model's architecture by isolating its components, with results indicating that the global network is particularly effective in this task. In conclusion, MLG2Net exhibited promising applications in DRP for cancer cell lines, with potential advancements by incorporating larger datasets.
Collapse
Affiliation(s)
- Thi-Oanh Tran
- Hematology and Blood Transfusion Center, Bach Mai Hospital, Hanoi, Viet Nam
- AIBioMed Research Group, Taipei Medical University, Taipei, 110, Taiwan
| | - Thanh-Huy Nguyen
- AIBioMed Research Group, Taipei Medical University, Taipei, 110, Taiwan
- Saigon Precision Medicine Research Center, Ho Chi Minh city, Vietnam
| | - Tuan Tung Nguyen
- Hematology and Blood Transfusion Center, Bach Mai Hospital, Hanoi, Viet Nam
| | - Nguyen Quoc Khanh Le
- AIBioMed Research Group, Taipei Medical University, Taipei, 110, Taiwan.
- In-Service Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei, 110, Taiwan.
| |
Collapse
|
3
|
Wang C, Kumar GA, Rajapakse JC. Drug discovery and mechanism prediction with explainable graph neural networks. Sci Rep 2025; 15:179. [PMID: 39747341 PMCID: PMC11696803 DOI: 10.1038/s41598-024-83090-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 12/11/2024] [Indexed: 01/04/2025] Open
Abstract
Apprehension of drug action mechanism is paramount for drug response prediction and precision medicine. The unprecedented development of machine learning and deep learning algorithms has expedited the drug response prediction research. However, existing methods mainly focus on forward encoding of drugs, which is to obtain an accurate prediction of the response levels, but omitted to decipher the reaction mechanism between drug molecules and genes. We propose the eXplainable Graph-based Drug response Prediction (XGDP) approach that achieves a precise drug response prediction and reveals the comprehensive mechanism of action between drugs and their targets. XGDP represents drugs with molecular graphs, which naturally preserve the structural information of molecules and a Graph Neural Network module is applied to learn the latent features of molecules. Gene expression data from cancer cell lines are incorporated and processed by a Convolutional Neural Network module. A couple of deep learning attribution algorithms are leveraged to interpret interactions between drug molecular features and genes. We demonstrate that XGDP not only enhances the prediction accuracy compared to pioneering works but is also capable of capturing the salient functional groups of drugs and interactions with significant genes of cancer cells.
Collapse
Affiliation(s)
- Conghao Wang
- College of Computing and Data Science, Nanyang Technological University, Singapore, 639798, Singapore
| | - Gaurav Asok Kumar
- College of Computing and Data Science, Nanyang Technological University, Singapore, 639798, Singapore
| | - Jagath C Rajapakse
- College of Computing and Data Science, Nanyang Technological University, Singapore, 639798, Singapore.
| |
Collapse
|
4
|
Zhao Y, Yang K, Chen Y, Lv Z, Wang Q, Zhong Y, Chen X. Machine learning-based pan-cancer study of classification and mechanism of BRAF inhibitor resistance. Transl Cancer Res 2024; 13:6645-6660. [PMID: 39816555 PMCID: PMC11730697 DOI: 10.21037/tcr-24-961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/25/2024] [Indexed: 01/18/2025]
Abstract
Background V-raf murine sarcoma viral oncogene homolog B1 (BRAF) inhibitor (BRAFi) therapy resistance affects approximately 15% of cancer patients, leading to disease recurrence and poor prognosis. The aim of the study was to develop a machine-learning based method to identify patients who are at high-risk of BRAFi resistance and potential biomarker. Methods From Cancer Cell Line Encyclopedia (CCLE) and Genomics of Drug Sensitivity in Cancer (GDSC) databases, we collected RNA sequencing and half maximal inhibitory concentration (IC50) data from 235 pan-cancer cell lines and then identified 37 significant differential expression genes associated with BRAFi resistance. Employing machine learning (ML) models, we successfully classified cell lines into resistant and sensitive groups, achieving robust performance in external validation datasets. Results AOX1 may play a vital part in BRAFi metabolism and resistance. Further, we found that higher mRNA expression of OXTR, H2AC13, and TBX2, and lower mRNA of SLC2A4, as detected by PCR in WM983B and SKMEL-5 cell lines, were independent risk factors for BRAFi resistance and were associated with poor prognosis. Conclusions We established a gene-expression model using ML methods, consisting of 37 variables based on RNA-seq database, which was externally validated and could be used to predict BRAFi resistance. Meanwhile, our findings provide valuable insights into the molecular mechanisms of BRAFi resistance, enabling the identification of high-risk patients.
Collapse
Affiliation(s)
- Yuhang Zhao
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Kai Yang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yujun Chen
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Zexi Lv
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Qing Wang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuanyuan Zhong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiqun Chen
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Wang J, Wang X, Pang Y. StructNet-DDI: Molecular Structure Characterization-Based ResNet for Prediction of Drug-Drug Interactions. Molecules 2024; 29:4829. [PMID: 39459198 PMCID: PMC11510539 DOI: 10.3390/molecules29204829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/30/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
This study introduces a deep learning framework based on SMILES representations of chemical structures to predict drug-drug interactions (DDIs). The model extracts Morgan fingerprints and key molecular descriptors, transforming them into raw graphical features for input into a modified ResNet18 architecture. The deep residual network, enhanced with regularization techniques, efficiently addresses training issues such as gradient vanishing and exploding, resulting in superior predictive performance. Experimental results show that StructNet-DDI achieved an AUC of 99.7%, an accuracy of 94.4%, and an AUPR of 99.9%, demonstrating the model's effectiveness and reliability. These findings highlight that StructNet-DDI can effectively extract crucial features from molecular structures, offering a simple yet robust tool for DDI prediction.
Collapse
Affiliation(s)
- Jihong Wang
- School of Computer, Guangdong University of Education, Guangzhou 510310, China
| | - Xiaodan Wang
- School of Pharmaceutical Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China
| | - Yuyao Pang
- School of Pharmaceutical Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China
| |
Collapse
|
6
|
Xia X, Zhu C, Zhong F, Liu L. TransCDR: a deep learning model for enhancing the generalizability of drug activity prediction through transfer learning and multimodal data fusion. BMC Biol 2024; 22:227. [PMID: 39385185 PMCID: PMC11462810 DOI: 10.1186/s12915-024-02023-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 09/30/2024] [Indexed: 10/11/2024] Open
Abstract
BACKGROUND Accurate and robust drug response prediction is of utmost importance in precision medicine. Although many models have been developed to utilize the representations of drugs and cancer cell lines for predicting cancer drug responses (CDR), their performances can be improved by addressing issues such as insufficient data modality, suboptimal fusion algorithms, and poor generalizability for novel drugs or cell lines. RESULTS We introduce TransCDR, which uses transfer learning to learn drug representations and fuses multi-modality features of drugs and cell lines by a self-attention mechanism, to predict the IC50 values or sensitive states of drugs on cell lines. We are the first to systematically evaluate the generalization of the CDR prediction model to novel (i.e., never-before-seen) compound scaffolds and cell line clusters. TransCDR shows better generalizability than 8 state-of-the-art models. TransCDR outperforms its 5 variants that train drug encoders (i.e., RNN and AttentiveFP) from scratch under various scenarios. The most critical contributors among multiple drug notations and omics profiles are Extended Connectivity Fingerprint and genetic mutation. Additionally, the attention-based fusion module further enhances the predictive performance of TransCDR. TransCDR, trained on the GDSC dataset, demonstrates strong predictive performance on the external testing set CCLE. It is also utilized to predict missing CDRs on GDSC. Moreover, we investigate the biological mechanisms underlying drug response by classifying 7675 patients from TCGA into drug-sensitive or drug-resistant groups, followed by a Gene Set Enrichment Analysis. CONCLUSIONS TransCDR emerges as a potent tool with significant potential in drug response prediction.
Collapse
Affiliation(s)
- Xiaoqiong Xia
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Chaoyu Zhu
- Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China
| | - Fan Zhong
- Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China.
| | - Lei Liu
- Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China.
| |
Collapse
|
7
|
Sharma A, Lysenko A, Jia S, Boroevich KA, Tsunoda T. Advances in AI and machine learning for predictive medicine. J Hum Genet 2024; 69:487-497. [PMID: 38424184 PMCID: PMC11422165 DOI: 10.1038/s10038-024-01231-y] [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: 10/31/2023] [Revised: 02/04/2024] [Accepted: 02/12/2024] [Indexed: 03/02/2024]
Abstract
The field of omics, driven by advances in high-throughput sequencing, faces a data explosion. This abundance of data offers unprecedented opportunities for predictive modeling in precision medicine, but also presents formidable challenges in data analysis and interpretation. Traditional machine learning (ML) techniques have been partly successful in generating predictive models for omics analysis but exhibit limitations in handling potential relationships within the data for more accurate prediction. This review explores a revolutionary shift in predictive modeling through the application of deep learning (DL), specifically convolutional neural networks (CNNs). Using transformation methods such as DeepInsight, omics data with independent variables in tabular (table-like, including vector) form can be turned into image-like representations, enabling CNNs to capture latent features effectively. This approach not only enhances predictive power but also leverages transfer learning, reducing computational time, and improving performance. However, integrating CNNs in predictive omics data analysis is not without challenges, including issues related to model interpretability, data heterogeneity, and data size. Addressing these challenges requires a multidisciplinary approach, involving collaborations between ML experts, bioinformatics researchers, biologists, and medical doctors. This review illuminates these complexities and charts a course for future research to unlock the full predictive potential of CNNs in omics data analysis and related fields.
Collapse
Affiliation(s)
- Alok Sharma
- Laboratory for Medical Science Mathematics, Department of Biological Sciences, School of Science, The University of Tokyo, Tokyo, Japan.
- Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- Institute for Integrated and Intelligent Systems, Griffith University, Queensland, Australia.
| | - Artem Lysenko
- Laboratory for Medical Science Mathematics, Department of Biological Sciences, School of Science, The University of Tokyo, Tokyo, Japan.
- Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
| | - Shangru Jia
- Laboratory for Medical Science Mathematics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Keith A Boroevich
- Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Tatsuhiko Tsunoda
- Laboratory for Medical Science Mathematics, Department of Biological Sciences, School of Science, The University of Tokyo, Tokyo, Japan.
- Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- Laboratory for Medical Science Mathematics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
8
|
Tan Z, Zhao Y, Lin K, Zhou T. Multi-task pretrained language model with novel application domains enables more comprehensive health and ecological toxicity prediction. JOURNAL OF HAZARDOUS MATERIALS 2024; 477:135265. [PMID: 39038381 DOI: 10.1016/j.jhazmat.2024.135265] [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: 04/13/2024] [Revised: 06/29/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
In silico models for screening substances of healthy and ecological concern are essential for effective chemical management. However, current data-driven toxicity prediction models confront formidable challenges related to expressive capacity, data scarcity, and reliability issues. Thus, this study introduces TOX-BERT, a SMILES-based pretrained model for screening health and ecological toxicity. Results show that masked atom recovery pretraining and multi-task learning offer promising solutions to enhance model capacity and address data scarcity issues. Two novel application domain (AD) parameters, termed PCA-AD and LDS, were proposed to improve prediction reliability of TOX-BERT with accuracy surpassing 90 % and mean absolute error (MAE) below 0.52. TOX-BERT was applied to 18,905 IECSC chemicals, revealing distinct toxicity relationships that align with experimental studies such as those between cardiotoxicity and acute ecotoxicity. In addition to previous PBT screening, 156 potential high-risk chemicals for specific endpoint were identified covering 7 categories. Furthermore, a SMILES-based toxicity site detection approach was developed for structural toxicity analysis. These advancements carry profound implications to address challenges faced by current data-driven toxicity prediction models. TOX-BERT emerges as a valuable tool for more comprehensive, reliable, and applicable predictions of health and ecological toxicity in chemical risk assessment and management.
Collapse
Affiliation(s)
- Zhichao Tan
- The State Key Laboratory of Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai 200092, China; Shanghai Institute of Pollution Control and Ecological Security, 1515 North Zhongshan Rd. (No. 2), Shanghai 200092, PR China.
| | - Youcai Zhao
- The State Key Laboratory of Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai 200092, China; Shanghai Institute of Pollution Control and Ecological Security, 1515 North Zhongshan Rd. (No. 2), Shanghai 200092, PR China.
| | - Kunsen Lin
- The State Key Laboratory of Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai 200092, China; Shanghai Institute of Pollution Control and Ecological Security, 1515 North Zhongshan Rd. (No. 2), Shanghai 200092, PR China.
| | - Tao Zhou
- The State Key Laboratory of Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai 200092, China; Shanghai Institute of Pollution Control and Ecological Security, 1515 North Zhongshan Rd. (No. 2), Shanghai 200092, PR China.
| |
Collapse
|
9
|
Li B, Tan K, Lao AR, Wang H, Zheng H, Zhang L. A comprehensive review of artificial intelligence for pharmacology research. Front Genet 2024; 15:1450529. [PMID: 39290983 PMCID: PMC11405247 DOI: 10.3389/fgene.2024.1450529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024] Open
Abstract
With the innovation and advancement of artificial intelligence, more and more artificial intelligence techniques are employed in drug research, biomedical frontier research, and clinical medicine practice, especially, in the field of pharmacology research. Thus, this review focuses on the applications of artificial intelligence in drug discovery, compound pharmacokinetic prediction, and clinical pharmacology. We briefly introduced the basic knowledge and development of artificial intelligence, presented a comprehensive review, and then summarized the latest studies and discussed the strengths and limitations of artificial intelligence models. Additionally, we highlighted several important studies and pointed out possible research directions.
Collapse
Affiliation(s)
- Bing Li
- College of Computer Science, Sichuan University, Chengdu, China
| | - Kan Tan
- College of Computer Science, Sichuan University, Chengdu, China
| | - Angelyn R Lao
- Department of Mathematics and Statistics, De La Salle University, Manila, Philippines
| | - Haiying Wang
- School of Computing, Ulster University, Belfast, United Kingdom
| | - Huiru Zheng
- School of Computing, Ulster University, Belfast, United Kingdom
| | - Le Zhang
- College of Computer Science, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Sun YY, Hsieh CY, Wen JH, Tseng TY, Huang JH, Oyang YJ, Huang HC, Juan HF. scDrug+: predicting drug-responses using single-cell transcriptomics and molecular structure. Biomed Pharmacother 2024; 177:117070. [PMID: 38964180 DOI: 10.1016/j.biopha.2024.117070] [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: 04/19/2024] [Revised: 06/18/2024] [Accepted: 06/29/2024] [Indexed: 07/06/2024] Open
Abstract
Predicting drug responses based on individual transcriptomic profiles holds promise for refining prognosis and advancing precision medicine. Although many studies have endeavored to predict the responses of known drugs to novel transcriptomic profiles, research into predicting responses for newly discovered drugs remains sparse. In this study, we introduce scDrug+, a comprehensive pipeline that seamlessly integrates single-cell analysis with drug-response prediction. Importantly, scDrug+ is equipped to predict the response of new drugs by analyzing their molecular structures. The open-source tool is available as a Docker container, ensuring ease of deployment and reproducibility. It can be accessed at https://github.com/ailabstw/scDrugplus.
Collapse
Affiliation(s)
- Yih-Yun Sun
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taiwan; Taiwan AI Labs, Taipei 10351, Taiwan
| | | | - Jian-Hung Wen
- Taiwan AI Labs, Taipei 10351, Taiwan; Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Tzu-Yang Tseng
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taiwan; Department of Life Science, National Taiwan University, Taipei 106, Taiwan
| | | | - Yen-Jen Oyang
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taiwan
| | - Hsuan-Cheng Huang
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan.
| | - Hsueh-Fen Juan
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taiwan; Taiwan AI Labs, Taipei 10351, Taiwan; Department of Life Science, National Taiwan University, Taipei 106, Taiwan; Center for Computational and Systems Biology, National Taiwan University, Taipei 106, Taiwan; Center for Advanced Computing and Imaging in Biomedicine, National Taiwan University, Taipei 106, Taiwan.
| |
Collapse
|
11
|
Eckhart L, Lenhof K, Rolli LM, Lenhof HP. A comprehensive benchmarking of machine learning algorithms and dimensionality reduction methods for drug sensitivity prediction. Brief Bioinform 2024; 25:bbae242. [PMID: 38797968 PMCID: PMC11128483 DOI: 10.1093/bib/bbae242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 04/05/2024] [Accepted: 05/06/2024] [Indexed: 05/29/2024] Open
Abstract
A major challenge of precision oncology is the identification and prioritization of suitable treatment options based on molecular biomarkers of the considered tumor. In pursuit of this goal, large cancer cell line panels have successfully been studied to elucidate the relationship between cellular features and treatment response. Due to the high dimensionality of these datasets, machine learning (ML) is commonly used for their analysis. However, choosing a suitable algorithm and set of input features can be challenging. We performed a comprehensive benchmarking of ML methods and dimension reduction (DR) techniques for predicting drug response metrics. Using the Genomics of Drug Sensitivity in Cancer cell line panel, we trained random forests, neural networks, boosting trees and elastic nets for 179 anti-cancer compounds with feature sets derived from nine DR approaches. We compare the results regarding statistical performance, runtime and interpretability. Additionally, we provide strategies for assessing model performance compared with a simple baseline model and measuring the trade-off between models of different complexity. Lastly, we show that complex ML models benefit from using an optimized DR strategy, and that standard models-even when using considerably fewer features-can still be superior in performance.
Collapse
Affiliation(s)
- Lea Eckhart
- Center for Bioinformatics, Saarland Informatics Campus, Saarland University, 66123, Saarland, Germany
| | - Kerstin Lenhof
- Center for Bioinformatics, Saarland Informatics Campus, Saarland University, 66123, Saarland, Germany
| | - Lisa-Marie Rolli
- Center for Bioinformatics, Saarland Informatics Campus, Saarland University, 66123, Saarland, Germany
| | - Hans-Peter Lenhof
- Center for Bioinformatics, Saarland Informatics Campus, Saarland University, 66123, Saarland, Germany
| |
Collapse
|
12
|
Inoue Y, Lee H, Fu T, Luna A. drGAT: Attention-Guided Gene Assessment of Drug Response Utilizing a Drug-Cell-Gene Heterogeneous Network. ARXIV 2024:arXiv:2405.08979v1. [PMID: 38800657 PMCID: PMC11118660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Drug development is a lengthy process with a high failure rate. Increasingly, machine learning is utilized to facilitate the drug development processes. These models aim to enhance our understanding of drug characteristics, including their activity in biological contexts. However, a major challenge in drug response (DR) prediction is model interpretability as it aids in the validation of findings. This is important in biomedicine, where models need to be understandable in comparison with established knowledge of drug interactions with proteins. drGAT, a graph deep learning model, leverages a heterogeneous graph composed of relationships between proteins, cell lines, and drugs. drGAT is designed with two objectives: DR prediction as a binary sensitivity prediction and elucidation of drug mechanism from attention coefficients. drGAT has demonstrated superior performance over existing models, achieving 78% accuracy (and precision), and 76% F1 score for 269 DNA-damaging compounds of the NCI60 drug response dataset. To assess the model's interpretability, we conducted a review of drug-gene co-occurrences in Pubmed abstracts in comparison to the top 5 genes with the highest attention coefficients for each drug. We also examined whether known relationships were retained in the model by inspecting the neighborhoods of topoisomerase-related drugs. For example, our model retained TOP1 as a highly weighted predictive feature for irinotecan and topotecan, in addition to other genes that could potentially be regulators of the drugs. Our method can be used to accurately predict sensitivity to drugs and may be useful in the identification of biomarkers relating to the treatment of cancer patients.
Collapse
Affiliation(s)
- Yoshitaka Inoue
- Department of Computer Science and Engineering, University of Minnesota
- Computational Biology Branch, National Library of Medicine
| | - Hunmin Lee
- Department of Computer Science and Engineering, University of Minnesota
| | - Tianfan Fu
- Computer Science Department, Rensselaer Polytechnic Institute
| | - Augustin Luna
- Computational Biology Branch, National Library of Medicine
- Developmental Therapeutics Branch, National Cancer Institute
| |
Collapse
|
13
|
Pang Y, Chen Y, Lin M, Zhang Y, Zhang J, Wang L. MMSyn: A New Multimodal Deep Learning Framework for Enhanced Prediction of Synergistic Drug Combinations. J Chem Inf Model 2024; 64:3689-3705. [PMID: 38676916 DOI: 10.1021/acs.jcim.4c00165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2024]
Abstract
Combination therapy is a promising strategy for the successful treatment of cancer. The large number of possible combinations, however, mean that it is laborious and expensive to screen for synergistic drug combinations in vitro. Nevertheless, because of the availability of high-throughput screening data and advances in computational techniques, deep learning (DL) can be a useful tool for the prediction of synergistic drug combinations. In this study, we proposed a multimodal DL framework, MMSyn, for the prediction of synergistic drug combinations. First, features embedded in the drug molecules were extracted: structure, fingerprint, and string encoding. Then, gene expression data, DNA copy number, and pathway activity were used to describe cancer cell lines. Finally, these processed features were integrated using an attention mechanism and an interaction module and then input into a multilayer perceptron to predict drug synergy. Experimental results showed that our method outperformed five state-of-the-art DL methods and three traditional machine learning models for drug combination prediction. We verified that MMSyn achieved superior performance in stratified cross-validation settings using both the drug combination and cell line data. Moreover, we performed a set of ablation experiments to illustrate the effectiveness of each component and the efficacy of our model. In addition, our visual representation and case studies further confirmed the effectiveness of our model. All results showed that MMSyn can be used as a powerful tool for the prediction of synergistic drug combinations.
Collapse
Affiliation(s)
- Yu Pang
- Joint International Research Laboratory of Synthetic Biology and Medicine, Ministry of Education, Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Yihao Chen
- Joint International Research Laboratory of Synthetic Biology and Medicine, Ministry of Education, Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Mujie Lin
- Joint International Research Laboratory of Synthetic Biology and Medicine, Ministry of Education, Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Yanhong Zhang
- Joint International Research Laboratory of Synthetic Biology and Medicine, Ministry of Education, Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Jiquan Zhang
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, College of Pharmacy, Guizhou Medical University, Guiyang 550025, P. R. China
| | - Ling Wang
- Joint International Research Laboratory of Synthetic Biology and Medicine, Ministry of Education, Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
14
|
Jiang J, Li Y, Zhang R, Liu Y. INTransformer: Data augmentation-based contrastive learning by injecting noise into transformer for molecular property prediction. J Mol Graph Model 2024; 128:108703. [PMID: 38228013 DOI: 10.1016/j.jmgm.2024.108703] [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: 10/10/2023] [Revised: 12/05/2023] [Accepted: 01/02/2024] [Indexed: 01/18/2024]
Abstract
Molecular property prediction plays an essential role in drug discovery for identifying the candidate molecules with target properties. Deep learning models usually require sufficient labeled data to train good prediction models. However, the size of labeled data is usually small for molecular property prediction, which brings great challenges to deep learning-based molecular property prediction methods. Furthermore, the global information of molecules is critical for predicting molecular properties. Therefore, we propose INTransformer for molecular property prediction, which is a data augmentation method via contrastive learning to alleviate the limitations of the labeled molecular data while enhancing the ability to capture global information. Specifically, INTransformer consists of two identical Transformer sub-encoders to extract the molecular representation from the original SMILES and noisy SMILES respectively, while achieving the goal of data augmentation. To reduce the influence of noise, we use contrastive learning to ensure the molecular encoding of noisy SMILES is consistent with that of the original input so that the molecular representation information can be better extracted by INTransformer. Experiments on various benchmark datasets show that INTransformer achieved competitive performance for molecular property prediction tasks compared with the baselines and state-of-the-art methods.
Collapse
Affiliation(s)
- Jing Jiang
- Key Laboratory of Linguistic and Cultural Computing, Ministry of Education, Northwest Minzu University, Lanzhou 730030, China.
| | - Yachao Li
- Key Laboratory of Linguistic and Cultural Computing, Ministry of Education, Northwest Minzu University, Lanzhou 730030, China.
| | - Ruisheng Zhang
- School of Information Science and Engineering, Lanzhou University, Lanzhou 730000, China.
| | - Yunwu Liu
- School of Information Science and Engineering, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
15
|
Lin CX, Guan Y, Li HD. Artificial intelligence approaches for molecular representation in drug response prediction. Curr Opin Struct Biol 2024; 84:102747. [PMID: 38091924 DOI: 10.1016/j.sbi.2023.102747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/26/2023] [Accepted: 11/26/2023] [Indexed: 02/09/2024]
Abstract
Drug response prediction is essential for drug development and disease treatment. One key question in predicting drug response is the representation of molecules, which has been greatly advanced by artificial intelligence (AI) techniques in recent years. In this review, we first describe different types of representation methods, pinpointing their key principles and discussing their limitations. Thereafter we discuss potential ways how these methods could be further developed. We expect that this review will provide useful guidance for researchers in the community.
Collapse
Affiliation(s)
- Cui-Xiang Lin
- School of Mathematics and Computational Science, Xiangtan University, Xiangtan, 411105, Hunan Province, PR China
| | - Yuanfang Guan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Hong-Dong Li
- School of Computer Science and Engineering, Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha, Hunan 410083, PR China.
| |
Collapse
|
16
|
Tan Z, Zhao Y, Zhou T, Lin K. Hi-MGT: A hybrid molecule graph transformer for toxicity identification. JOURNAL OF HAZARDOUS MATERIALS 2023; 457:131808. [PMID: 37307723 DOI: 10.1016/j.jhazmat.2023.131808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/18/2023] [Accepted: 06/07/2023] [Indexed: 06/14/2023]
Abstract
Conventional toxicity testing methods that rely on animal experimentation are resource-intensive, time-consuming, and ethically controversial. Therefore, the development of alternative non-animal testing approaches is crucial. This study proposes a novel hybrid graph transformer architecture, termed Hi-MGT, for the toxicity identification. An innovative aggregation strategy, referred to as GNN-GT combination, enables Hi-MGT to simultaneously and comprehensively aggregate local and global structural information of molecules, thus elucidating more informative toxicity information hidden in molecule graphs. The results show that the state-of-the-art model outperforms current baseline CML and DL models on a diverse range of toxicity endpoints and is even comparable to large-scale pretrained GNNs with geometry enhancement. Additionally, the impact of hyperparameters on model performance is investigated, and a systematic ablation study is conducted to demonstrate the effectiveness of the GNN-GT combination. Moreover, this study provides valuable insights into the learning process on molecules and proposes a novel similarity-based method for toxic site detection, which could potentially facilitate toxicity identification and analysis. Overall, the Hi-MGT model represents a significant advancement in the development of alternative non-animal testing approaches for toxicity identification, with promising implications for enhancing human safety in the use of chemical compounds.
Collapse
Affiliation(s)
- Zhichao Tan
- The State Key Laboratory of Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai 200092, China; Shanghai Institute of Pollution Control and Ecological Security, 1515 North Zhongshan Rd. (No. 2), Shanghai 200092, China
| | - Youcai Zhao
- The State Key Laboratory of Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai 200092, China; Shanghai Institute of Pollution Control and Ecological Security, 1515 North Zhongshan Rd. (No. 2), Shanghai 200092, China
| | - Tao Zhou
- The State Key Laboratory of Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai 200092, China; Shanghai Institute of Pollution Control and Ecological Security, 1515 North Zhongshan Rd. (No. 2), Shanghai 200092, China.
| | - Kunsen Lin
- The State Key Laboratory of Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai 200092, China; Shanghai Institute of Pollution Control and Ecological Security, 1515 North Zhongshan Rd. (No. 2), Shanghai 200092, China.
| |
Collapse
|
17
|
Zhang M, Gao H, Liao X, Ning B, Gu H, Yu B. DBGRU-SE: predicting drug-drug interactions based on double BiGRU and squeeze-and-excitation attention mechanism. Brief Bioinform 2023:7176312. [PMID: 37225428 DOI: 10.1093/bib/bbad184] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/03/2023] [Accepted: 04/23/2023] [Indexed: 05/26/2023] Open
Abstract
The prediction of drug-drug interactions (DDIs) is essential for the development and repositioning of new drugs. Meanwhile, they play a vital role in the fields of biopharmaceuticals, disease diagnosis and pharmacological treatment. This article proposes a new method called DBGRU-SE for predicting DDIs. Firstly, FP3 fingerprints, MACCS fingerprints, Pubchem fingerprints and 1D and 2D molecular descriptors are used to extract the feature information of the drugs. Secondly, Group Lasso is used to remove redundant features. Then, SMOTE-ENN is applied to balance the data to obtain the best feature vectors. Finally, the best feature vectors are fed into the classifier combining BiGRU and squeeze-and-excitation (SE) attention mechanisms to predict DDIs. After applying five-fold cross-validation, The ACC values of DBGRU-SE model on the two datasets are 97.51 and 94.98%, and the AUC are 99.60 and 98.85%, respectively. The results showed that DBGRU-SE had good predictive performance for drug-drug interactions.
Collapse
Affiliation(s)
| | - Hongli Gao
- Qingdao University of Science and Technology, China
| | - Xin Liao
- Qingdao University of Science and Technology, China
| | - Baoxing Ning
- Qingdao University of Science and Technology, China
| | - Haiming Gu
- Qingdao University of Science and Technology, China
| | - Bin Yu
- Qingdao University of Science and Technology, China
| |
Collapse
|
18
|
Kour S, Biswas I, Sheoran S, Arora S, Sheela P, Duppala SK, Murthy DK, Pawar SC, Singh H, Kumar D, Prabhu D, Vuree S, Kumar R. Artificial intelligence and nanotechnology for cervical cancer treatment: Current status and future perspectives. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
|
19
|
Partin A, Brettin TS, Zhu Y, Narykov O, Clyde A, Overbeek J, Stevens RL. Deep learning methods for drug response prediction in cancer: Predominant and emerging trends. Front Med (Lausanne) 2023; 10:1086097. [PMID: 36873878 PMCID: PMC9975164 DOI: 10.3389/fmed.2023.1086097] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/23/2023] [Indexed: 02/17/2023] Open
Abstract
Cancer claims millions of lives yearly worldwide. While many therapies have been made available in recent years, by in large cancer remains unsolved. Exploiting computational predictive models to study and treat cancer holds great promise in improving drug development and personalized design of treatment plans, ultimately suppressing tumors, alleviating suffering, and prolonging lives of patients. A wave of recent papers demonstrates promising results in predicting cancer response to drug treatments while utilizing deep learning methods. These papers investigate diverse data representations, neural network architectures, learning methodologies, and evaluations schemes. However, deciphering promising predominant and emerging trends is difficult due to the variety of explored methods and lack of standardized framework for comparing drug response prediction models. To obtain a comprehensive landscape of deep learning methods, we conducted an extensive search and analysis of deep learning models that predict the response to single drug treatments. A total of 61 deep learning-based models have been curated, and summary plots were generated. Based on the analysis, observable patterns and prevalence of methods have been revealed. This review allows to better understand the current state of the field and identify major challenges and promising solution paths.
Collapse
Affiliation(s)
- Alexander Partin
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
| | - Thomas S. Brettin
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
| | - Yitan Zhu
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
| | - Oleksandr Narykov
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
| | - Austin Clyde
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
| | - Jamie Overbeek
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
| | - Rick L. Stevens
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
- Department of Computer Science, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
20
|
Wang H, Dai C, Wen Y, Wang X, Liu W, He S, Bo X, Peng S. GADRP: graph convolutional networks and autoencoders for cancer drug response prediction. Brief Bioinform 2023; 24:6865039. [PMID: 36460622 DOI: 10.1093/bib/bbac501] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/19/2022] [Accepted: 10/22/2022] [Indexed: 12/04/2022] Open
Abstract
Drug response prediction in cancer cell lines is of great significance in personalized medicine. In this study, we propose GADRP, a cancer drug response prediction model based on graph convolutional networks (GCNs) and autoencoders (AEs). We first use a stacked deep AE to extract low-dimensional representations from cell line features, and then construct a sparse drug cell line pair (DCP) network incorporating drug, cell line, and DCP similarity information. Later, initial residual and layer attention-based GCN (ILGCN) that can alleviate over-smoothing problem is utilized to learn DCP features. And finally, fully connected network is employed to make prediction. Benchmarking results demonstrate that GADRP can significantly improve prediction performance on all metrics compared with baselines on five datasets. Particularly, experiments of predictions of unknown DCP responses, drug-cancer tissue associations, and drug-pathway associations illustrate the predictive power of GADRP. All results highlight the effectiveness of GADRP in predicting drug responses, and its potential value in guiding anti-cancer drug selection.
Collapse
Affiliation(s)
- Hong Wang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Chong Dai
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China.,Department of Bioinformatics, Beijing Institute of Health Service and Transfusion Medicine, Beijing 100850, China
| | - Yuqi Wen
- Department of Bioinformatics, Beijing Institute of Health Service and Transfusion Medicine, Beijing 100850, China
| | - Xiaoqi Wang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Wenjuan Liu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Song He
- Department of Bioinformatics, Beijing Institute of Health Service and Transfusion Medicine, Beijing 100850, China
| | - Xiaochen Bo
- Department of Bioinformatics, Beijing Institute of Health Service and Transfusion Medicine, Beijing 100850, China
| | - Shaoliang Peng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China.,The State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha 410082, China
| |
Collapse
|
21
|
Shen B, Feng F, Li K, Lin P, Ma L, Li H. A systematic assessment of deep learning methods for drug response prediction: from in vitro to clinical applications. Brief Bioinform 2023; 24:6961794. [PMID: 36575826 DOI: 10.1093/bib/bbac605] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/30/2022] [Accepted: 12/09/2022] [Indexed: 12/29/2022] Open
Abstract
Drug response prediction is an important problem in personalized cancer therapy. Among various newly developed models, significant improvement in prediction performance has been reported using deep learning methods. However, systematic comparisons of deep learning methods, especially of the transferability from preclinical models to clinical cohorts, are currently lacking. To provide a more rigorous assessment, the performance of six representative deep learning methods for drug response prediction using nine evaluation metrics, including the overall prediction accuracy, predictability of each drug, potential associated factors and transferability to clinical cohorts, in multiple application scenarios was benchmarked. Most methods show promising prediction within cell line datasets, and TGSA, with its lower time cost and better performance, is recommended. Although the performance metrics decrease when applying models trained on cell lines to patients, a certain amount of power to distinguish clinical response on some drugs can be maintained using CRDNN and TGSA. With these assessments, we provide a guidance for researchers to choose appropriate methods, as well as insights into future directions for the development of more effective methods in clinical scenarios.
Collapse
Affiliation(s)
- Bihan Shen
- Cancer Systems Biology group at Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Fangyoumin Feng
- Cancer Systems Biology group at Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Kunshi Li
- Cancer Systems Biology group at Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ping Lin
- Cancer Systems Biology group at Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Liangxiao Ma
- Bio-Med Big Data Center at Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hong Li
- Cancer Systems Biology group at Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
22
|
Du BX, Qin Y, Jiang YF, Xu Y, Yiu SM, Yu H, Shi JY. Compound–protein interaction prediction by deep learning: Databases, descriptors and models. Drug Discov Today 2022; 27:1350-1366. [DOI: 10.1016/j.drudis.2022.02.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/19/2021] [Accepted: 02/28/2022] [Indexed: 11/24/2022]
|