1
|
Ren Z, Zeng X, Lao Y, You Z, Shang Y, Zou Q, Lin C. Predicting rare drug-drug interaction events with dual-granular structure-adaptive and pair variational representation. Nat Commun 2025; 16:3997. [PMID: 40301328 PMCID: PMC12041321 DOI: 10.1038/s41467-025-59431-9] [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: 08/13/2024] [Accepted: 04/16/2025] [Indexed: 05/01/2025] Open
Abstract
Adverse drug-drug interaction events (DDIEs) pose serious risks to patient safety, yet rare but severe interactions remain challenging to identify due to limited clinical data. Existing computational methods rely heavily on abundant samples, failing to identify rare DDIEs. Here we introduce RareDDIE, a metric-based meta-learning model that employs a dual-granular structure-driven pair variational representation to enhance rare DDIE prediction. To further address the challenge of zero-shot DDIE identification, we develop the Biological Semantic Transferring (BST) module, integrating large-scale sentence embeddings to form the ZetaDDIE variant. Our model outperforms existing methods in few-sample and zero-sample settings. Furthermore, we verify that knowledge transfer from DDIE can improve drug synergy predictions, surpassing existing models. Case studies on antiplatelet activity reduction and non-small cell lung cancer drug synergy further illustrate the practical value of RareDDIE. By analyzing the meta-knowledge construction process, we provide interpretability into the model's decision-making. This work establishes an effective computational framework for rare DDIE prediction, leveraging meta-learning and knowledge transfer to overcome key challenges in data-limited scenarios.
Collapse
Affiliation(s)
- Zhonghao Ren
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410082, China
| | - Xiangxiang Zeng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410082, China
| | - Yizhen Lao
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410082, China
| | - Zhuhong You
- China School of Computer Science, Northwestern Polytechnical University, Xi'an, 710129, China
| | - Yifan Shang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410082, China
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 610054, China.
| | - Chen Lin
- School of Informatics, National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, 361000, China.
- Zhongguancun Academy, Beijing, China.
| |
Collapse
|
2
|
Jiang W, Ye W, Tan X, Bao YJ. Network-based multi-omics integrative analysis methods in drug discovery: a systematic review. BioData Min 2025; 18:27. [PMID: 40155979 PMCID: PMC11954193 DOI: 10.1186/s13040-025-00442-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 03/17/2025] [Indexed: 04/01/2025] Open
Abstract
The integration of multi-omics data from diverse high-throughput technologies has revolutionized drug discovery. While various network-based methods have been developed to integrate multi-omics data, systematic evaluation and comparison of these methods remain challenging. This review aims to analyze network-based approaches for multi-omics integration and evaluate their applications in drug discovery. We conducted a comprehensive review of literature (2015-2024) on network-based multi-omics integration methods in drug discovery, and categorized methods into four primary types: network propagation/diffusion, similarity-based approaches, graph neural networks, and network inference models. We also discussed the applications of the methods in three scenario of drug discovery, including drug target identification, drug response prediction, and drug repurposing, and finally evaluated the performance of the methods by highlighting their advantages and limitations in specific applications. While network-based multi-omics integration has shown promise in drug discovery, challenges remain in computational scalability, data integration, and biological interpretation. Future developments should focus on incorporating temporal and spatial dynamics, improving model interpretability, and establishing standardized evaluation frameworks.
Collapse
Affiliation(s)
- Wei Jiang
- School of Life Sciences, Hubei University, Wuhan, China
| | - Weicai Ye
- School of Computer Science and Engineering, Guangdong Province Key Laboratory of Computational Science, National Engineering Laboratory for Big Data Analysis and Application, Sun Yat-sen University, Guangzhou, China
| | - Xiaoming Tan
- School of Life Sciences, Hubei University, Wuhan, China
| | - Yun-Juan Bao
- School of Life Sciences, Hubei University, Wuhan, China.
- , No.368 Youyi Avenue, Wuhan, 430062, China.
| |
Collapse
|
3
|
Wang X, Zhu H, Liu Q, Liu Q. Fusing Micro- and Macro-Scale Information to Predict Anticancer Synergistic Drug Combinations. IEEE J Biomed Health Inform 2025; 29:2297-2309. [PMID: 40030241 DOI: 10.1109/jbhi.2024.3500789] [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: 03/08/2025]
Abstract
Drug combination therapy is highly regarded in cancer treatment. Computational methods offer a time- and cost-effective opportunity to explore the vast combination space. Although deep learning-based prediction methods lead the field, their generalization ability remains unsatisfactory. Few previous studies have the ability to finely characterize drugs and cell lines at both the micro-scale and macro-scale. Furthermore, the interaction of cross-scale information is often overlooked. These two points limit models' ability of predicting the synergism of drug combinations in cell lines. To address the issues, we propose a novel anticancer synergistic drug combination prediction method termed MMFSynergy in this article. The construction of MMFSynergy involves three phases. First, MMFSynergy pretrains two micro encoders and a macro graph encoder, which can capture micro- or macro-scale information from large volumes of unlabeled data and generate generic features for drugs and proteins. Second, it represents drugs and proteins by fusing cross-scale information through a self-supervised task. Finally, it employs a Transformer Encoder-based model to predict synergy scores, taking representations of drugs in the combinations and the associated proteins of cell lines as input. We compared our method with eight advanced methods across three typical scenarios based on two public datasets. The results consistently demonstrated that the proposed method's generalization ability outperforms six advanced methods'. We also conducted experiments including but not limited to ablation study and case study to further exhibit the effectiveness of MMFSynergy.
Collapse
|
4
|
Chen J, Lin A, Jiang A, Qi C, Liu Z, Cheng Q, Yuan S, Luo P. Computational frameworks transform antagonism to synergy in optimizing combination therapies. NPJ Digit Med 2025; 8:44. [PMID: 39828791 PMCID: PMC11743742 DOI: 10.1038/s41746-025-01435-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025] Open
Abstract
While drug combinations are increasingly important in disease treatment, predicting their therapeutic interactions remains challenging. This review systematically analyzes computational methods for predicting drug combination effects through multi-omics data integration. We comprehensively assess key algorithms including DrugComboRanker and AuDNNsynergy, and evaluate integration approaches encompassing kernel regression and graph networks. The review elucidates artificial intelligence applications in predicting drug synergistic and antagonistic effects.
Collapse
Affiliation(s)
- Jinghong Chen
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Chang Qi
- Vienna University of Technology, Institute of Logic and Computation, Vienna, Austria
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Shuofeng Yuan
- Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China.
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong, Hong Kong, China.
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
5
|
Jiang Z, Ding P, Shen C, Dai X. Geometric Molecular Graph Representation Learning Model for Drug-Drug Interactions Prediction. IEEE J Biomed Health Inform 2024; 28:7623-7632. [PMID: 39226203 DOI: 10.1109/jbhi.2024.3453956] [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: 09/05/2024]
Abstract
Drug-drug interaction (DDI) can trigger many adverse effects in patients and has emerged as a threat to medicine and public health. Therefore, it is important to predict potential drug interactions since it can provide combination strategies of drugs for systematic and effective treatment. Existing deep learning-based methods often rely on DDI functional networks, or use them as an important part of the model information source. However, it is difficult to discover the interactions of a new drug. To address the above limitations, we propose a geometric molecular graph representation learning model (Mol-DDI) for DDI prediction based on the basic assumption that structure determines function. Mol-DDI only considers the covalent and non-covalent bond information of molecules, then it uses the pre-training idea of large-scale models to learn drug molecular representations and predict drug interactions during the fine-tuning process. Experimental results show that the Mol-DDI model outperforms others on the three datasets and performs better in predicting new drug interaction experiments.
Collapse
|
6
|
Rani P, Dutta K, Kumar V. Autoencoder-based drug synergy framework for malignant diseases. Comput Biol Chem 2024; 113:108273. [PMID: 39522484 DOI: 10.1016/j.compbiolchem.2024.108273] [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: 07/04/2024] [Revised: 10/21/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Drug combination emerges as a viable option for the treatment of malignant diseases. Drug combination outperforms monotherapy by improving therapeutic efficacy, reducing toxicity, and overcoming drug resistance. To find viable drug combinations it is difficult to traverse empirically because of enormous combinational space. Machine learning and deep learning approaches are used to uncover novel synergistic drug combinations in enormous combinational space. Here, AESyn, a novel autoencoder-based drug synergy framework for malignant diseases using a bag of words encoding is proposed. The bag of word encoding technique is used to extract drug-targeted genes. The framework utilized screening data from NCI-ALMANAC, and O'Neil datasets. Autoencoders take drug embeddings with drug-targeted genes as input for processing. The autoencoder in the proposed framework is used to extract drug features. The proposed framework is evaluated on classification and regression metrics. The performance of the proposed framework is compared with existing methods of drug synergy. According to the findings, the proposed framework achieved high performance with an accuracy of 95%, AUROC of 94.2%, and MAPE of 7.2. The autoencoder-based framework for malignant diseases using an encoding technique provides a stable, order-independent drug synergy prediction.
Collapse
Affiliation(s)
- Pooja Rani
- Computer Science and Engineering Department, National Institute of Technology, Hamirpur, HP, 177005, India
| | - Kamlesh Dutta
- Computer Science and Engineering Department, National Institute of Technology, Hamirpur, HP, 177005, India
| | - Vijay Kumar
- Information Technology Department, Dr. B R Ambedkar National Institute of Technology Jalandhar, Punjab, 144027, India.
| |
Collapse
|
7
|
Monem S, Hassanien AE, Abdel-Hamid AH. A multi-view feature representation for predicting drugs combination synergy based on ensemble and multi-task attention models. J Cheminform 2024; 16:110. [PMID: 39334437 PMCID: PMC11438216 DOI: 10.1186/s13321-024-00903-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024] Open
Abstract
This paper proposes a novel multi-view ensemble predictor model that is designed to address the challenge of determining synergistic drug combinations by predicting both the synergy score value values and synergy class label of drug combinations with cancer cell lines. The proposed methodology involves representing drug features through four distinct views: Simplified Molecular-Input Line-Entry System (SMILES) features, molecular graph features, fingerprint features, and drug-target features. On the other hand, cell line features are captured through four views: gene expression features, copy number features, mutation features, and proteomics features. To prevent overfitting of the model, two techniques are employed. First, each view feature of a drug is paired with each corresponding cell line view and input into a multi-task attention deep learning model. This multi-task model is trained to simultaneously predict both the synergy score value and synergy class label. This process results in sixteen input view features being fed into the multi-task model, producing sixteen prediction values. Subsequently, these prediction values are utilized as inputs for an ensemble model, which outputs the final prediction value. The 'MVME' model is assessed using the O'Neil dataset, which includes 38 distinct drugs combined across 39 distinct cancer cell lines to output 22,737 drug combination pairs. For the synergy score value, the proposed model scores a mean square error (MSE) of 206.57, a root mean square error (RMSE) of 14.30, and a Pearson score of 0.76. For the synergy class label, the model scores 0.90 for accuracy, 0.96 for precision, 0.57 for kappa, 0.96 for the area under the ROC curve (ROC-AUC), and 0.88 for the area under the precision-recall curve (PR-AUC).
Collapse
Affiliation(s)
- Samar Monem
- Mathematics and Computer Science Department, Faculty of Science, Beni-Suef University, Beni-Suef, 62521, Egypt.
| | | | - Alaa H Abdel-Hamid
- Mathematics and Computer Science Department, Faculty of Science, Beni-Suef University, Beni-Suef, 62521, Egypt
| |
Collapse
|
8
|
Yang T, Li H, Kang Y, Li Z. MMFSyn: A Multimodal Deep Learning Model for Predicting Anticancer Synergistic Drug Combination Effect. Biomolecules 2024; 14:1039. [PMID: 39199425 PMCID: PMC11352627 DOI: 10.3390/biom14081039] [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: 06/30/2024] [Revised: 08/10/2024] [Accepted: 08/16/2024] [Indexed: 09/01/2024] Open
Abstract
Combination therapy aims to synergistically enhance efficacy or reduce toxic side effects and has widely been used in clinical practice. However, with the rapid increase in the types of drug combinations, identifying the synergistic relationships between drugs remains a highly challenging task. This paper proposes a novel deep learning model MMFSyn based on multimodal drug data combined with cell line features. Firstly, to ensure the full expression of drug molecular features, multiple modalities of drugs, including Morgan fingerprints, atom sequences, molecular diagrams, and atomic point cloud data, are extracted using SMILES. Secondly, for different modal data, a Bi-LSTM, gMLP, multi-head attention mechanism, and multi-scale GCNs are comprehensively applied to extract the drug feature. Then, it selects appropriate omics features from gene expression and mutation omics data of cancer cell lines to construct cancer cell line features. Finally, these features are combined to predict the synergistic anti-cancer drug combination effect. The experimental results verify that MMFSyn has significant advantages in performance compared to other popular methods, with a root mean square error of 13.33 and a Pearson correlation coefficient of 0.81, which indicates that MMFSyn can better capture the complex relationship between multimodal drug combinations and omics data, thereby improving the synergistic drug combination prediction.
Collapse
Affiliation(s)
- Tao Yang
- School of Information Engineering, Huzhou University, Huzhou 313000, China;
- College of Science, Zhejiang Sci-Tech University, Hangzhou 310018, China;
| | - Haohao Li
- College of Science, Zhejiang Sci-Tech University, Hangzhou 310018, China;
| | - Yanlei Kang
- School of Information Engineering, Huzhou University, Huzhou 313000, China;
| | - Zhong Li
- School of Information Engineering, Huzhou University, Huzhou 313000, China;
- College of Science, Zhejiang Sci-Tech University, Hangzhou 310018, China;
| |
Collapse
|
9
|
Alidoost M, Wilson JL. Preclinical side effect prediction through pathway engineering of protein interaction network models. CPT Pharmacometrics Syst Pharmacol 2024; 13:1180-1200. [PMID: 38736280 PMCID: PMC11247120 DOI: 10.1002/psp4.13150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/01/2024] [Accepted: 04/08/2024] [Indexed: 05/14/2024] Open
Abstract
Modeling tools aim to predict potential drug side effects, although they suffer from imperfect performance. Specifically, protein-protein interaction models predict drug effects from proteins surrounding drug targets, but they tend to overpredict drug phenotypes and require well-defined pathway phenotypes. In this study, we used PathFX, a protein-protein interaction tool, to predict side effects for active ingredient-side effect pairs extracted from drug labels. We observed limited performance and defined new pathway phenotypes using pathway engineering strategies. We defined new pathway phenotypes using a network-based and gene expression-based approach. Overall, we discovered a trade-off between sensitivity and specificity values and demonstrated a way to limit overprediction for side effects with sufficient true positive examples. We compared our predictions to animal models and demonstrated similar performance metrics, suggesting that protein-protein interaction models do not need perfect evaluation metrics to be useful. Pathway engineering, through the inclusion of true positive examples and omics measurements, emerges as a promising approach to enhance the utility of protein interaction network models for drug effect prediction.
Collapse
Affiliation(s)
- Mohammadali Alidoost
- Department of Bioengineering, University of California, Los Angeles, California, USA
| | - Jennifer L Wilson
- Department of Bioengineering, University of California, Los Angeles, California, USA
| |
Collapse
|
10
|
Dong Y, Bai Y, Liu H, Yang Z, Chang Y, Li J, Han Q, Feng X, Fan X, Ren X. DKPE-GraphSYN: a drug synergy prediction model based on joint dual kernel density estimation and positional encoding for graph representation. Front Genet 2024; 15:1401544. [PMID: 38948360 PMCID: PMC11211516 DOI: 10.3389/fgene.2024.1401544] [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: 03/15/2024] [Accepted: 05/24/2024] [Indexed: 07/02/2024] Open
Abstract
Introduction: Synergistic medication, a crucial therapeutic strategy in cancer treatment, involves combining multiple drugs to enhance therapeutic effectiveness and mitigate side effects. Current research predominantly employs deep learning models for extracting features from cell line and cancer drug structure data. However, these methods often overlook the intricate nonlinear relationships within the data, neglecting the distribution characteristics and weighted probability densities of gene expression data in multi-dimensional space. It also fails to fully exploit the structural information of cancer drugs and the potential interactions between drug molecules. Methods: To overcome these challenges, we introduce an innovative end-to-end learning model specifically tailored for cancer drugs, named Dual Kernel Density and Positional Encoding (DKPE) for Graph Synergy Representation Network (DKPEGraphSYN). This model is engineered to refine the prediction of drug combination synergy effects in cancer. DKPE-GraphSYN utilizes Dual Kernel Density Estimation and Positional Encoding techniques to effectively capture the weighted probability density and spatial distribution information of gene expression, while exploring the interactions and potential relationships between cancer drug molecules via a graph neural network. Results: Experimental results show that our prediction model achieves significant performance enhancements in forecasting drug synergy effects on a comprehensive cancer drug and cell line synergy dataset, achieving an AUPR of 0.969 and an AUC of 0.976. Discussion: These results confirm our model's superior accuracy in predicting cancer drug combinations, providing a supportive method for clinical medication strategy in cancer.
Collapse
Affiliation(s)
- Yunyun Dong
- School of Software, Taiyuan University of Technology, Taiyuan, Shanxi, China
| | - Yujie Bai
- School of Software, Taiyuan University of Technology, Taiyuan, Shanxi, China
| | - Haitao Liu
- School of Software, Taiyuan University of Technology, Taiyuan, Shanxi, China
| | - Ziting Yang
- School of Software, Taiyuan University of Technology, Taiyuan, Shanxi, China
| | - Yunqing Chang
- School of Software, Taiyuan University of Technology, Taiyuan, Shanxi, China
| | - Jianguang Li
- School of Software, Taiyuan University of Technology, Taiyuan, Shanxi, China
| | - Qixuan Han
- School of Software, Taiyuan University of Technology, Taiyuan, Shanxi, China
| | - Xiufang Feng
- School of Software, Taiyuan University of Technology, Taiyuan, Shanxi, China
| | - Xiaole Fan
- Information Management Department, Shanxi Provincial People’s Hospital, Taiyuan, Shanxi, China
| | - Xiaoqiang Ren
- Information Management Department, Shanxi Provincial People’s Hospital, Taiyuan, Shanxi, China
| |
Collapse
|
11
|
Huangfu X, Zhang C, Li H, Li S, Li Y. SNSynergy: Similarity network-based machine learning framework for synergy prediction towards new cell lines and new anticancer drug combinations. Comput Biol Chem 2024; 110:108054. [PMID: 38522389 DOI: 10.1016/j.compbiolchem.2024.108054] [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: 10/25/2023] [Revised: 02/22/2024] [Accepted: 03/16/2024] [Indexed: 03/26/2024]
Abstract
The computational method has been proven to be a promising means for pre-screening large-scale anticancer drug combinations to support precision oncology applications. Pioneering efforts have been made to develop machine learning technology for predicting drug synergy, but high computational cost for training models as well as great diversity and limited size in screening data escalate the difficulty of prediction. To address this challenge, we propose a simple machine learning framework, namely Similarity Network-based Synergy prediction (SNSynergy), for predicting synergistic effects towards new cell lines and new drug combinations by two locally weighted models CLSN and DCSN. This framework only requires a small amount of auxiliary data, like genomics information of cell lines and the molecular fingerprints or targets of drugs. Based on the assumption that similar cell lines and similar drug combinations have similar synergistic effects, CLSN and DCSN predict synergy scores through capturing individual synergy contributions of nearest cell line and drug combination neighbors, respectively. High correlations between predicted and measured synergy scores on two leading cancer cell line pharmacogenomic screening datasets (the O'Neil dataset and the NCI-ALMANAC dataset) demonstrate the effectiveness and robustness of SNSynergy. Many of the identified drug combinations are consistent with previous studies, or have been explored in clinical settings against the specific cancer type, showing that SNSynergy has the potential to supply cost-saving and effective high-throughput screening for prioritizing the most applicable cell lines and the most promising drug combinations.
Collapse
Affiliation(s)
- Xiaosheng Huangfu
- School of Science, Yanshan University, Qinhuangdao, Hebei 066004, China
| | - Chengwei Zhang
- School of Science, Yanshan University, Qinhuangdao, Hebei 066004, China
| | - Hualong Li
- School of Science, Yanshan University, Qinhuangdao, Hebei 066004, China; School of mathematics and statistics, Guangdong University of Technology, Guangzhou 510520, China
| | - Sile Li
- School of Science, Yanshan University, Qinhuangdao, Hebei 066004, China
| | - Yushuang Li
- School of Science, Yanshan University, Qinhuangdao, Hebei 066004, China.
| |
Collapse
|
12
|
Cheng N, Wang L, Liu Y, Song B, Ding C. HANSynergy: Heterogeneous Graph Attention Network for Drug Synergy Prediction. J Chem Inf Model 2024; 64:4334-4347. [PMID: 38709204 PMCID: PMC11135324 DOI: 10.1021/acs.jcim.4c00003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/07/2024]
Abstract
Drug synergy therapy is a promising strategy for cancer treatment. However, the extensive variety of available drugs and the time-intensive process of determining effective drug combinations through clinical trials pose significant challenges. It requires a reliable method for the rapid and precise selection of drug synergies. In response, various computational strategies have been developed for predicting drug synergies, yet the exploitation of heterogeneous biological network features remains underexplored. In this study, we construct a heterogeneous graph that encompasses diverse biological entities and interactions, utilizing rich data sets from sources, such as DrugCombDB, PubChem, UniProt, and cancer cell line encyclopedia (CCLE). We initialize node feature representations and introduce a novel virtual node to enhance drug representation. Our proposed method, the heterogeneous graph attention network for drug-drug synergy prediction (HANSynergy), has been experimentally validated to demonstrate that the heterogeneous graph attention network can extract key node features, efficiently harness the diversity of information, and further enhance network functionality through the incorporation of a multihead attention mechanism. In the comparative experiment, the highest accuracy (Acc) and area under the curve (AUC) are 0.877 and 0.947, respectively, in DrugCombDB_early data set, demonstrating the superiority of HANSynergy over the competing methods. Moreover, protein-protein interactions are important in understanding the mechanism of action of drugs. The heterogeneous attention mechanism facilitates protein-protein interaction analysis. By analyzing the changes of attention weight before and after heterogeneous network training, we investigated proteins that may be associated with drug combinations. Additionally, case studies align our findings with existing research, underscoring the potential of HANSynergy in drug synergy prediction. This advancement not only contributes to the burgeoning field of drug synergy prediction but also holds the potential to provide valuable insights and uncover new drug synergies for combating cancer.
Collapse
Affiliation(s)
- Ning Cheng
- School
of Informatics, Hunan University of Chinese
Medicine, Changsha, Hunan 410208, China
| | - Li Wang
- Degree
Programs in Systems and information Engineering, University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan
| | - Yiping Liu
- College
of Information Science and Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Bosheng Song
- College
of Information Science and Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Changsong Ding
- School
of Informatics, Hunan University of Chinese
Medicine, Changsha, Hunan 410208, China
- Big
Data Analysis Laboratory of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| |
Collapse
|
13
|
Wang S, Qiu Y, Zhu F. An updated review of functional ingredients of Manuka honey and their value-added innovations. Food Chem 2024; 440:138060. [PMID: 38211407 DOI: 10.1016/j.foodchem.2023.138060] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 11/19/2023] [Accepted: 11/20/2023] [Indexed: 01/13/2024]
Abstract
Manuka honey (MH) is a highly prized natural product from the nectar of Leptospermum scoparium flowers. Increased competition on the global market drives MH product innovations. This review updates comparative and non-comparative studies to highlight nutritional, therapeutic, bioengineering, and cosmetic values of MH. MH is a good source of phenolics and unique chemical compounds, such as methylglyoxal, dihydroxyacetone, leptosperin glyoxal, methylsyringate and leptosin. Based on the evidence from in vitro, in vivo and clinical studies, multifunctional bioactive compounds of MH have exhibited anti-oxidative, anti-inflammatory, immunomodulatory, anti-microbial, and anti-cancer activities. There are controversial topics related to MH, such as MH grading, safety/efficacy, implied benefits, and maximum levels of contaminants concerned. Artificial intelligence can optimize MH studies related to chemical analysis, toxicity prediction, multi-functional mechanism exploration and product innovation.
Collapse
Affiliation(s)
- Sunan Wang
- Canadian Food and Wine Institute, Niagara College, 135 Taylor Road, Niagara-on-the-Lake, Ontario L0S 1J0, Canada; School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Yi Qiu
- Division of Engineering Science, Faculty of Applied Science and Engineering, University of Toronto, 35 St. George Street, Toronto, Ontario M5S 1A4, Canada
| | - Fan Zhu
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| |
Collapse
|
14
|
Liu X, Abad L, Chatterjee L, Cristea IM, Varjosalo M. Mapping protein-protein interactions by mass spectrometry. MASS SPECTROMETRY REVIEWS 2024:10.1002/mas.21887. [PMID: 38742660 PMCID: PMC11561166 DOI: 10.1002/mas.21887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 04/22/2024] [Indexed: 05/16/2024]
Abstract
Protein-protein interactions (PPIs) are essential for numerous biological activities, including signal transduction, transcription control, and metabolism. They play a pivotal role in the organization and function of the proteome, and their perturbation is associated with various diseases, such as cancer, neurodegeneration, and infectious diseases. Recent advances in mass spectrometry (MS)-based protein interactomics have significantly expanded our understanding of the PPIs in cells, with techniques that continue to improve in terms of sensitivity, and specificity providing new opportunities for the study of PPIs in diverse biological systems. These techniques differ depending on the type of interaction being studied, with each approach having its set of advantages, disadvantages, and applicability. This review highlights recent advances in enrichment methodologies for interactomes before MS analysis and compares their unique features and specifications. It emphasizes prospects for further improvement and their potential applications in advancing our knowledge of PPIs in various biological contexts.
Collapse
Affiliation(s)
- Xiaonan Liu
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
- Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Lawrence Abad
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Lopamudra Chatterjee
- Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Ileana M. Cristea
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Markku Varjosalo
- Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| |
Collapse
|
15
|
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
|
16
|
Dong Y, Chang Y, Wang Y, Han Q, Wen X, Yang Z, Zhang Y, Qiang Y, Wu K, Fan X, Ren X. MFSynDCP: multi-source feature collaborative interactive learning for drug combination synergy prediction. BMC Bioinformatics 2024; 25:140. [PMID: 38561679 PMCID: PMC10985899 DOI: 10.1186/s12859-024-05765-y] [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: 01/17/2024] [Accepted: 03/28/2024] [Indexed: 04/04/2024] Open
Abstract
Drug combination therapy is generally more effective than monotherapy in the field of cancer treatment. However, screening for effective synergistic combinations from a wide range of drug combinations is particularly important given the increase in the number of available drug classes and potential drug-drug interactions. Existing methods for predicting the synergistic effects of drug combinations primarily focus on extracting structural features of drug molecules and cell lines, but neglect the interaction mechanisms between cell lines and drug combinations. Consequently, there is a deficiency in comprehensive understanding of the synergistic effects of drug combinations. To address this issue, we propose a drug combination synergy prediction model based on multi-source feature interaction learning, named MFSynDCP, aiming to predict the synergistic effects of anti-tumor drug combinations. This model includes a graph aggregation module with an adaptive attention mechanism for learning drug interactions and a multi-source feature interaction learning controller for managing information transfer between different data sources, accommodating both drug and cell line features. Comparative studies with benchmark datasets demonstrate MFSynDCP's superiority over existing methods. Additionally, its adaptive attention mechanism graph aggregation module identifies drug chemical substructures crucial to the synergy mechanism. Overall, MFSynDCP is a robust tool for predicting synergistic drug combinations. The source code is available from GitHub at https://github.com/kkioplkg/MFSynDCP .
Collapse
Affiliation(s)
- Yunyun Dong
- School of Software, Taiyuan University of Technology, Taiyuan, Shanxi, China.
| | - Yunqing Chang
- School of Software, Taiyuan University of Technology, Taiyuan, Shanxi, China
| | - Yuxiang Wang
- School of Software, Taiyuan University of Technology, Taiyuan, Shanxi, China
| | - Qixuan Han
- School of Software, Taiyuan University of Technology, Taiyuan, Shanxi, China
| | - Xiaoyuan Wen
- School of Software, Taiyuan University of Technology, Taiyuan, Shanxi, China
| | - Ziting Yang
- School of Software, Taiyuan University of Technology, Taiyuan, Shanxi, China
| | - Yan Zhang
- School of Software, Taiyuan University of Technology, Taiyuan, Shanxi, China
| | - Yan Qiang
- College of Computer Science and Technology (College of Data Science), Taiyuan University of Technology, Taiyuan, Shanxi, China.
| | - Kun Wu
- School of Computing, University of Leeds, Leeds, West Yorkshire, UK
| | - Xiaole Fan
- Information Management Department, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Xiaoqiang Ren
- Information Management Department, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| |
Collapse
|
17
|
Yan S, Zheng D. A Deep Neural Network for Predicting Synergistic Drug Combinations on Cancer. Interdiscip Sci 2024; 16:218-230. [PMID: 38183569 DOI: 10.1007/s12539-023-00596-6] [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: 08/13/2023] [Revised: 11/19/2023] [Accepted: 11/21/2023] [Indexed: 01/08/2024]
Abstract
The exploration of drug combinations presents an opportunity to amplify therapeutic effectiveness while alleviating undesirable side effects. Nevertheless, the extensive array of potential combinations poses challenges in terms of cost and time constraints for experimental screening. Thus, it is crucial to narrow down the search space. Deep learning approaches have gained widespread popularity in predicting synergistic drug combinations tailored for specific cell lines in vitro settings. In the present study, we introduce a novel method termed GTextSyn, which utilizes the integration of gene expression data and chemical structure information for the prediction of synergistic effects in drug combinations. GTextSyn employs a sentence classification model within the domain of Natural Language Processing (NLP), wherein drugs and cell lines are regarded as entities possessing biochemical relevance. Meanwhile, combinations of drug pairs and cell lines are construed as sentences with biochemical relational significance. To assess the efficacy of GTextSyn, we conduct a comparative analysis with alternative deep learning approaches using a standard benchmark dataset. The results from a five-fold cross-validation demonstrate a 49.5% reduction in Mean Square Error (MSE) achieved by GTextSyn, surpassing the performance of the next best method in the regression task. Furthermore, we conduct a comprehensive literature survey on the predicted novel drug combinations and find substantial support from prior experimental studies for many of the combinations identified by GTextSyn.
Collapse
Affiliation(s)
- Shiyu Yan
- School of Computer, University of South China, West Changsheng Road, Hengyang, 421001, Hunan, China.
| | - Ding Zheng
- School of Computer, University of South China, West Changsheng Road, Hengyang, 421001, Hunan, China
| |
Collapse
|
18
|
Liu M, Srivastava G, Ramanujam J, Brylinski M. SynerGNet: A Graph Neural Network Model to Predict Anticancer Drug Synergy. Biomolecules 2024; 14:253. [PMID: 38540674 PMCID: PMC10967862 DOI: 10.3390/biom14030253] [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: 12/24/2023] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 01/03/2025] Open
Abstract
Drug combination therapy shows promise in cancer treatment by addressing drug resistance, reducing toxicity, and enhancing therapeutic efficacy. However, the intricate and dynamic nature of biological systems makes identifying potential synergistic drugs a costly and time-consuming endeavor. To facilitate the development of combination therapy, techniques employing artificial intelligence have emerged as a transformative solution, providing a sophisticated avenue for advancing existing therapeutic approaches. In this study, we developed SynerGNet, a graph neural network model designed to accurately predict the synergistic effect of drug pairs against cancer cell lines. SynerGNet utilizes cancer-specific featured graphs created by integrating heterogeneous biological features into the human protein-protein interaction network, followed by a reduction process to enhance topological diversity. Leveraging synergy data provided by AZ-DREAM Challenges, the model yields a balanced accuracy of 0.68, significantly outperforming traditional machine learning. Encouragingly, augmenting the training data with carefully constructed synthetic instances improved the balanced accuracy of SynerGNet to 0.73. Finally, the results of an independent validation conducted against DrugCombDB demonstrated that it exhibits a strong performance when applied to unseen data. SynerGNet shows a great potential in detecting drug synergy, positioning itself as a valuable tool that could contribute to the advancement of combination therapy for cancer treatment.
Collapse
Affiliation(s)
- Mengmeng Liu
- Division of Electrical and Computer Engineering, Louisiana State University, Baton Rouge, LA 70803, USA; (M.L.)
| | - Gopal Srivastava
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - J. Ramanujam
- Division of Electrical and Computer Engineering, Louisiana State University, Baton Rouge, LA 70803, USA; (M.L.)
- Center for Computation and Technology, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Michal Brylinski
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
- Center for Computation and Technology, Louisiana State University, Baton Rouge, LA 70803, USA
| |
Collapse
|
19
|
Kim DN, McNaughton AD, Kumar N. Leveraging Artificial Intelligence to Expedite Antibody Design and Enhance Antibody-Antigen Interactions. Bioengineering (Basel) 2024; 11:185. [PMID: 38391671 PMCID: PMC10886287 DOI: 10.3390/bioengineering11020185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
This perspective sheds light on the transformative impact of recent computational advancements in the field of protein therapeutics, with a particular focus on the design and development of antibodies. Cutting-edge computational methods have revolutionized our understanding of protein-protein interactions (PPIs), enhancing the efficacy of protein therapeutics in preclinical and clinical settings. Central to these advancements is the application of machine learning and deep learning, which offers unprecedented insights into the intricate mechanisms of PPIs and facilitates precise control over protein functions. Despite these advancements, the complex structural nuances of antibodies pose ongoing challenges in their design and optimization. Our review provides a comprehensive exploration of the latest deep learning approaches, including language models and diffusion techniques, and their role in surmounting these challenges. We also present a critical analysis of these methods, offering insights to drive further progress in this rapidly evolving field. The paper includes practical recommendations for the application of these computational techniques, supplemented with independent benchmark studies. These studies focus on key performance metrics such as accuracy and the ease of program execution, providing a valuable resource for researchers engaged in antibody design and development. Through this detailed perspective, we aim to contribute to the advancement of antibody design, equipping researchers with the tools and knowledge to navigate the complexities of this field.
Collapse
Affiliation(s)
| | | | - Neeraj Kumar
- Pacific Northwest National Laboratory, 902 Battelle Blvd., Richland, WA 99352, USA; (D.N.K.); (A.D.M.)
| |
Collapse
|
20
|
Guo Y, Hu H, Chen W, Yin H, Wu J, Hsieh CY, He Q, Cao J. SynergyX: a multi-modality mutual attention network for interpretable drug synergy prediction. Brief Bioinform 2024; 25:bbae015. [PMID: 38340091 PMCID: PMC10858681 DOI: 10.1093/bib/bbae015] [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: 09/24/2023] [Revised: 12/18/2023] [Indexed: 02/12/2024] Open
Abstract
Discovering effective anti-tumor drug combinations is crucial for advancing cancer therapy. Taking full account of intricate biological interactions is highly important in accurately predicting drug synergy. However, the extremely limited prior knowledge poses great challenges in developing current computational methods. To address this, we introduce SynergyX, a multi-modality mutual attention network to improve anti-tumor drug synergy prediction. It dynamically captures cross-modal interactions, allowing for the modeling of complex biological networks and drug interactions. A convolution-augmented attention structure is adopted to integrate multi-omic data in this framework effectively. Compared with other state-of-the-art models, SynergyX demonstrates superior predictive accuracy in both the General Test and Blind Test and cross-dataset validation. By exhaustively screening combinations of approved drugs, SynergyX reveals its ability to identify promising drug combination candidates for potential lung cancer treatment. Another notable advantage lies in its multidimensional interpretability. Taking Sorafenib and Vorinostat as an example, SynergyX serves as a powerful tool for uncovering drug-gene interactions and deciphering cell selectivity mechanisms. In summary, SynergyX provides an illuminating and interpretable framework, poised to catalyze the expedition of drug synergy discovery and deepen our comprehension of rational combination therapy.
Collapse
Affiliation(s)
- Yue Guo
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
| | - Haitao Hu
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
- Polytechnic Institute, Zhejiang University, 269 Shixiang Road,310000, Hangzhou, Zhejiang, China
| | - Wenbo Chen
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
- Polytechnic Institute, Zhejiang University, 269 Shixiang Road,310000, Hangzhou, Zhejiang, China
| | - Hao Yin
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
- Polytechnic Institute, Zhejiang University, 269 Shixiang Road,310000, Hangzhou, Zhejiang, China
| | - Jian Wu
- Second Affiliated Hospital School of Medicine, School of Public Health, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
| | - Chang-Yu Hsieh
- The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, 291 Fucheng Road, 310018, Hangzhou, Zhejiang, China
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
| | - Qiaojun He
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, 310020, Hangzhou, Zhejiang, China
- The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, 291 Fucheng Road, 310018, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
| | - Ji Cao
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, 310020, Hangzhou, Zhejiang, China
- The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, 291 Fucheng Road, 310018, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
| |
Collapse
|
21
|
Monem S, Hassanien AE, Abdel-Hamid AH. A multi-task learning model for predicting drugs combination synergy by analyzing drug-drug interactions and integrated multi-view graph data. Sci Rep 2023; 13:22463. [PMID: 38105262 PMCID: PMC10725868 DOI: 10.1038/s41598-023-48991-9] [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: 09/14/2023] [Accepted: 12/02/2023] [Indexed: 12/19/2023] Open
Abstract
This paper proposes a multi-task deep learning model for determining drug combination synergistic by simultaneously output synergy scores and synergy class labels. Initially, the two drugs are represented using a Simplified Molecular-Input Line-Entry (SMILE) system. Chemical structural features of the drugs are extracted from the SMILE using the RedKit package. Additionally, an improved Multi-view representation is proposed to extract graph-based drug features. Furthermore, the cancer cell line is represented by gene expression. Then, a three fully connected layers are learned to extract cancer cell line features. To investigate the impact of drug interactions on cell lines, the drug interaction features are extracted from a pretrained drugs interaction network and fed into an attention mechanism along with the cancer cell line features, resulting in the output of affected cancer cell line features. Subsequently, the drug and cell line features are concatenated and fed into an attention mechanism, which produces a two-feature representation for the two predicted tasks. The relationship between the two tasks is learned using the cross-stitch algorithm. Finally, each task feature is inputted into a fully connected subnetwork to predict the synergy score and synergy label. The proposed model 'MutliSyn' is evaluated using the O'Neil cancer dataset, comprising 38 unique drugs combined to form 22,737 drug combination pairs, tested on 39 cancer cell lines. For the synergy score, the model achieves a mean square error (MSE) of 219.14, a root mean square error (RMSE) of 14.75, and a Pearson score of 0.76. Regarding the synergy class label, the model achieves an area under the ROC curve (ROC-AUC) of 0.95, an area under the precision-recall curve (PR-AUC) of 0.85, precision of 0.93, kappa of 0.61, and accuracy of 0.90.
Collapse
Affiliation(s)
- Samar Monem
- Mathematics and Computer Science Department, Faculty of Science, Beni-Suef University, Beni-Suef, 62521, Egypt.
- Scientific Research Group in Egypt (SRGE), , .
| | - Aboul Ella Hassanien
- Faculty of Computer and AI, Cairo University, Cairo, Egypt
- Scientific Research Group in Egypt (SRGE),
| | - Alaa H Abdel-Hamid
- Mathematics and Computer Science Department, Faculty of Science, Beni-Suef University, Beni-Suef, 62521, Egypt
| |
Collapse
|
22
|
Chen D, Wang X, Zhu H, Jiang Y, Li Y, Liu Q, Liu Q. Predicting anticancer synergistic drug combinations based on multi-task learning. BMC Bioinformatics 2023; 24:448. [PMID: 38012551 PMCID: PMC10680313 DOI: 10.1186/s12859-023-05524-5] [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: 04/21/2023] [Accepted: 10/09/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND The discovery of anticancer drug combinations is a crucial work of anticancer treatment. In recent years, pre-screening drug combinations with synergistic effects in a large-scale search space adopting computational methods, especially deep learning methods, is increasingly popular with researchers. Although achievements have been made to predict anticancer synergistic drug combinations based on deep learning, the application of multi-task learning in this field is relatively rare. The successful practice of multi-task learning in various fields shows that it can effectively learn multiple tasks jointly and improve the performance of all the tasks. METHODS In this paper, we propose MTLSynergy which is based on multi-task learning and deep neural networks to predict synergistic anticancer drug combinations. It simultaneously learns two crucial prediction tasks in anticancer treatment, which are synergy prediction of drug combinations and sensitivity prediction of monotherapy. And MTLSynergy integrates the classification and regression of prediction tasks into the same model. Moreover, autoencoders are employed to reduce the dimensions of input features. RESULTS Compared with the previous methods listed in this paper, MTLSynergy achieves the lowest mean square error of 216.47 and the highest Pearson correlation coefficient of 0.76 on the drug synergy prediction task. On the corresponding classification task, the area under the receiver operator characteristics curve and the area under the precision-recall curve are 0.90 and 0.62, respectively, which are equivalent to the comparison methods. Through the ablation study, we verify that multi-task learning and autoencoder both have a positive effect on prediction performance. In addition, the prediction results of MTLSynergy in many cases are also consistent with previous studies. CONCLUSION Our study suggests that multi-task learning is significantly beneficial for both drug synergy prediction and monotherapy sensitivity prediction when combining these two tasks into one model. The ability of MTLSynergy to discover new anticancer synergistic drug combinations noteworthily outperforms other state-of-the-art methods. MTLSynergy promises to be a powerful tool to pre-screen anticancer synergistic drug combinations.
Collapse
Affiliation(s)
- Danyi Chen
- School of Software Engineering, Tongji University, Shanghai, 201804, China
| | - Xiaowen Wang
- School of Software Engineering, Tongji University, Shanghai, 201804, China
| | - Hongming Zhu
- School of Software Engineering, Tongji University, Shanghai, 201804, China
| | - Yizhi Jiang
- School of Software Engineering, Tongji University, Shanghai, 201804, China
| | - Yulong Li
- School of Software Engineering, Tongji University, Shanghai, 201804, China
| | - Qi Liu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Qin Liu
- School of Software Engineering, Tongji University, Shanghai, 201804, China.
| |
Collapse
|
23
|
Gan Y, Huang X, Guo W, Yan C, Zou G. Predicting synergistic anticancer drug combination based on low-rank global attention mechanism and bilinear predictor. Bioinformatics 2023; 39:btad607. [PMID: 37812255 PMCID: PMC10598574 DOI: 10.1093/bioinformatics/btad607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 08/29/2023] [Accepted: 10/07/2023] [Indexed: 10/10/2023] Open
Abstract
MOTIVATION Drug combination therapy has exhibited remarkable therapeutic efficacy and has gradually become a promising clinical treatment strategy of complex diseases such as cancers. As the related databases keep expanding, computational methods based on deep learning model have become powerful tools to predict synergistic drug combinations. However, predicting effective synergistic drug combinations is still a challenge due to the high complexity of drug combinations, the lack of biological interpretability, and the large discrepancy in the response of drug combinations in vivo and in vitro biological systems. RESULTS Here, we propose DGSSynADR, a new deep learning method based on global structured features of drugs and targets for predicting synergistic anticancer drug combinations. DGSSynADR constructs a heterogeneous graph by integrating the drug-drug, drug-target, protein-protein interactions and multi-omics data, utilizes a low-rank global attention (LRGA) model to perform global weighted aggregation of graph nodes and learn the global structured features of drugs and targets, and then feeds the embedded features into a bilinear predictor to predict the synergy scores of drug combinations in different cancer cell lines. Specifically, LRGA network brings better model generalization ability, and effectively reduces the complexity of graph computation. The bilinear predictor facilitates the dimension transformation of the features and fuses the feature representation of the two drugs to improve the prediction performance. The loss function Smooth L1 effectively avoids gradient explosion, contributing to better model convergence. To validate the performance of DGSSynADR, we compare it with seven competitive methods. The comparison results demonstrate that DGSSynADR achieves better performance. Meanwhile, the prediction of DGSSynADR is validated by previous findings in case studies. Furthermore, detailed ablation studies indicate that the one-hot coding drug feature, LRGA model and bilinear predictor play a key role in improving the prediction performance. AVAILABILITY AND IMPLEMENTATION DGSSynADR is implemented in Python using the Pytorch machine-learning library, and it is freely available at https://github.com/DHUDBlab/DGSSynADR.
Collapse
Affiliation(s)
- Yanglan Gan
- School of Computer Science and Technology, Donghua University, Shanghai 201600, China
| | - Xingyu Huang
- School of Computer Science and Technology, Donghua University, Shanghai 201600, China
| | - Wenjing Guo
- School of Computer Science and Technology, Donghua University, Shanghai 201600, China
| | - Cairong Yan
- School of Computer Science and Technology, Donghua University, Shanghai 201600, China
| | - Guobing Zou
- School of Computer Engineering and Science, Shanghai University, Shanghai 200444, China
| |
Collapse
|
24
|
Gu J, Bang D, Yi J, Lee S, Kim DK, Kim S. A model-agnostic framework to enhance knowledge graph-based drug combination prediction with drug-drug interaction data and supervised contrastive learning. Brief Bioinform 2023; 24:bbad285. [PMID: 37544660 DOI: 10.1093/bib/bbad285] [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: 05/15/2023] [Revised: 07/05/2023] [Accepted: 07/21/2023] [Indexed: 08/08/2023] Open
Abstract
Combination therapies have brought significant advancements to the treatment of various diseases in the medical field. However, searching for effective drug combinations remains a major challenge due to the vast number of possible combinations. Biomedical knowledge graph (KG)-based methods have shown potential in predicting effective combinations for wide spectrum of diseases, but the lack of credible negative samples has limited the prediction performance of machine learning models. To address this issue, we propose a novel model-agnostic framework that leverages existing drug-drug interaction (DDI) data as a reliable negative dataset and employs supervised contrastive learning (SCL) to transform drug embedding vectors to be more suitable for drug combination prediction. We conducted extensive experiments using various network embedding algorithms, including random walk and graph neural networks, on a biomedical KG. Our framework significantly improved performance metrics compared to the baseline framework. We also provide embedding space visualizations and case studies that demonstrate the effectiveness of our approach. This work highlights the potential of using DDI data and SCL in finding tighter decision boundaries for predicting effective drug combinations.
Collapse
Affiliation(s)
- Jeonghyeon Gu
- Interdisciplinary Program in Artificial Intelligence, Seoul National University, 1, Gwanak-ro, 08826 Seoul, Republic of Korea
| | - Dongmin Bang
- Interdisciplinary Program in Bioinformatics, Seoul National University, 1, Gwanak-ro, 08826 Seoul, Republic of Korea
- AIGENDRUG Co., Ltd., 1, Gwanak-ro, 08826 Seoul, Republic of Korea
| | - Jungseob Yi
- Interdisciplinary Program in Artificial Intelligence, Seoul National University, 1, Gwanak-ro, 08826 Seoul, Republic of Korea
| | - Sangseon Lee
- Institute of Computer Technology Seoul National University, 1, Gwanak-ro, 08826 Seoul, Republic of Korea
| | - Dong Kyu Kim
- PHARMGENSCIENCE Co., Ltd., 216, Dongjak-daero, 06554 Seoul, Republic of Korea
| | - Sun Kim
- Interdisciplinary Program in Artificial Intelligence, Seoul National University, 1, Gwanak-ro, 08826 Seoul, Republic of Korea
- Interdisciplinary Program in Bioinformatics, Seoul National University, 1, Gwanak-ro, 08826 Seoul, Republic of Korea
- Department of Computer Science and Engineering, Seoul National University, 1, Gwanak-ro, 08826 Seoul, Republic of Korea
- AIGENDRUG Co., Ltd., 1, Gwanak-ro, 08826 Seoul, Republic of Korea
- Institute of Computer Technology, Seoul National University, 1, Gwanak-ro, 08826 Seoul, Republic of Korea
| |
Collapse
|
25
|
Zhang P, Tu S. MGAE-DC: Predicting the synergistic effects of drug combinations through multi-channel graph autoencoders. PLoS Comput Biol 2023; 19:e1010951. [PMID: 36867661 PMCID: PMC10027223 DOI: 10.1371/journal.pcbi.1010951] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 03/20/2023] [Accepted: 02/14/2023] [Indexed: 03/04/2023] Open
Abstract
Accurate prediction of synergistic effects of drug combinations can reduce the experimental costs for drug development and facilitate the discovery of novel efficacious combination therapies for clinical studies. The drug combinations with high synergy scores are regarded as synergistic ones, while those with moderate or low synergy scores are additive or antagonistic ones. The existing methods usually exploit the synergy data from the aspect of synergistic drug combinations, paying little attention to the additive or antagonistic ones. Also, they usually do not leverage the common patterns of drug combinations across different cell lines. In this paper, we propose a multi-channel graph autoencoder (MGAE)-based method for predicting the synergistic effects of drug combinations (DC), and shortly denote it as MGAE-DC. A MGAE model is built to learn the drug embeddings by considering not only synergistic combinations but also additive and antagonistic ones as three input channels. The later two channels guide the model to explicitly characterize the features of non-synergistic combinations through an encoder-decoder learning process, and thus the drug embeddings become more discriminative between synergistic and non-synergistic combinations. In addition, an attention mechanism is incorporated to fuse each cell-line's drug embeddings across various cell lines, and a common drug embedding is extracted to capture the invariant patterns by developing a set of cell-line shared decoders. The generalization performance of our model is further improved with the invariant patterns. With the cell-line specific and common drug embeddings, our method is extended to predict the synergy scores of drug combinations by a neural network module. Experiments on four benchmark datasets demonstrate that MGAE-DC consistently outperforms the state-of-the-art methods. In-depth literature survey is conducted to find that many drug combinations predicted by MGAE-DC are supported by previous experimental studies. The source code and data are available at https://github.com/yushenshashen/MGAE-DC.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Computer Science and Engineering, Center for Cognitive Machines and Computational Health (CMaCH), Shanghai Jiao Tong University, Shanghai, China
| | - Shikui Tu
- Department of Computer Science and Engineering, Center for Cognitive Machines and Computational Health (CMaCH), Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
26
|
Li J, Li L, You P, Wei Y, Xu B. Towards artificial intelligence to multi-omics characterization of tumor heterogeneity in esophageal cancer. Semin Cancer Biol 2023; 91:35-49. [PMID: 36868394 DOI: 10.1016/j.semcancer.2023.02.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/21/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023]
Abstract
Esophageal cancer is a unique and complex heterogeneous malignancy, with substantial tumor heterogeneity: at the cellular levels, tumors are composed of tumor and stromal cellular components; at the genetic levels, they comprise genetically distinct tumor clones; at the phenotypic levels, cells in distinct microenvironmental niches acquire diverse phenotypic features. This heterogeneity affects almost every process of esophageal cancer progression from onset to metastases and recurrence, etc. Intertumoral and intratumoral heterogeneity are major obstacles in the treatment of esophageal cancer, but also offer the potential to manipulate the heterogeneity themselves as a new therapeutic strategy. The high-dimensional, multi-faceted characterization of genomics, epigenomics, transcriptomics, proteomics, metabonomics, etc. of esophageal cancer has opened novel horizons for dissecting tumor heterogeneity. Artificial intelligence especially machine learning and deep learning algorithms, are able to make decisive interpretations of data from multi-omics layers. To date, artificial intelligence has emerged as a promising computational tool for analyzing and dissecting esophageal patient-specific multi-omics data. This review provides a comprehensive review of tumor heterogeneity from a multi-omics perspective. Especially, we discuss the novel techniques single-cell sequencing and spatial transcriptomics, which have revolutionized our understanding of the cell compositions of esophageal cancer and allowed us to determine novel cell types. We focus on the latest advances in artificial intelligence in integrating multi-omics data of esophageal cancer. Artificial intelligence-based multi-omics data integration computational tools exert a key role in tumor heterogeneity assessment, which will potentially boost the development of precision oncology in esophageal cancer.
Collapse
Affiliation(s)
- Junyu Li
- Department of Radiation Oncology, Jiangxi Cancer Hospital, Nanchang 330029, Jiangxi, China; Jiangxi Health Committee Key (JHCK) Laboratory of Tumor Metastasis, Jiangxi Cancer Hospital, Nanchang 330029, Jiangxi, China
| | - Lin Li
- Department of Thoracic Oncology, Jiangxi Cancer Hospital, Nanchang 330029, Jiangxi, China
| | - Peimeng You
- Nanchang University, Department of Radiation Oncology, Jiangxi Cancer Hospital, Nanchang 330029, Jiangxi, China
| | - Yiping Wei
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China.
| | - Bin Xu
- Jiangxi Health Committee Key (JHCK) Laboratory of Tumor Metastasis, Jiangxi Cancer Hospital, Nanchang 330029, Jiangxi, China.
| |
Collapse
|
27
|
Lee M, Shin H, Park M, Kim A, Cha S, Lee H. Systems pharmacology approaches in herbal medicine research: a brief review. BMB Rep 2022; 55:417-428. [PMID: 35880436 PMCID: PMC9537023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/29/2022] [Accepted: 07/21/2022] [Indexed: 02/21/2025] Open
Abstract
Herbal medicine, a multi-component treatment, has been extensively practiced for treating various symptoms and diseases. However, its molecular mechanism of action on the human body is unknown, which impedes the development and application of herbal medicine. To address this, recent studies are increasingly adopting systems pharmacology, which interprets pharmacological effects of drugs from consequences of the interaction networks that drugs might have. Most conventional network- based approaches collect associations of herb-compound, compound-target, and target-disease from individual databases, respectively, and construct an integrated network of herb-compound- target-disease to study the complex mechanisms underlying herbal treatment. More recently, rapid advances in highthroughput omics technology have led numerous studies to exploring gene expression profiles induced by herbal treatments to elicit information on direct associations between herbs and genes at the genome-wide scale. In this review, we summarize key databases and computational methods utilized in systems pharmacology for studying herbal medicine. We also highlight recent studies that identify modes of action or novel indications of herbal medicine by harnessing drug-induced transcriptome data. [BMB Reports 2022; 55(9): 417-428].
Collapse
Affiliation(s)
- Myunggyo Lee
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Korea
| | - Hyejin Shin
- Korean Medicine (KM) Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea
| | - Musun Park
- Korean Medicine (KM) Data Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea
| | - Aeyung Kim
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Korea
| | - Seongwon Cha
- Korean Medicine (KM) Data Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea
| | - Haeseung Lee
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Korea
| |
Collapse
|
28
|
Rintala TJ, Ghosh A, Fortino V. Network approaches for modeling the effect of drugs and diseases. Brief Bioinform 2022; 23:6608969. [PMID: 35704883 PMCID: PMC9294412 DOI: 10.1093/bib/bbac229] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/29/2022] [Accepted: 05/17/2021] [Indexed: 12/12/2022] Open
Abstract
The network approach is quickly becoming a fundamental building block of computational methods aiming at elucidating the mechanism of action (MoA) and therapeutic effect of drugs. By modeling the effect of drugs and diseases on different biological networks, it is possible to better explain the interplay between disease perturbations and drug targets as well as how drug compounds induce favorable biological responses and/or adverse effects. Omics technologies have been extensively used to generate the data needed to study the mechanisms of action of drugs and diseases. These data are often exploited to define condition-specific networks and to study whether drugs can reverse disease perturbations. In this review, we describe network data mining algorithms that are commonly used to study drug’s MoA and to improve our understanding of the basis of chronic diseases. These methods can support fundamental stages of the drug development process, including the identification of putative drug targets, the in silico screening of drug compounds and drug combinations for the treatment of diseases. We also discuss recent studies using biological and omics-driven networks to search for possible repurposed FDA-approved drug treatments for SARS-CoV-2 infections (COVID-19).
Collapse
Affiliation(s)
- T J Rintala
- Institute of Biomedicine, University of Eastern Finland, 70210 Kuopio, Finland
| | - Arindam Ghosh
- Institute of Biomedicine, University of Eastern Finland, 70210 Kuopio, Finland
| | - V Fortino
- Institute of Biomedicine, University of Eastern Finland, 70210 Kuopio, Finland
| |
Collapse
|