1
|
Pourmousa M, Jain S, Barnaeva E, Jin W, Hochuli J, Itkin Z, Maxfield T, Melo-Filho C, Thieme A, Wilson K, Klumpp-Thomas C, Michael S, Southall N, Jaakkola T, Muratov EN, Barzilay R, Tropsha A, Ferrer M, Zakharov AV. AI-driven discovery of synergistic drug combinations against pancreatic cancer. Nat Commun 2025; 16:4020. [PMID: 40301300 PMCID: PMC12041571 DOI: 10.1038/s41467-025-56818-6] [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: 05/23/2024] [Accepted: 01/31/2025] [Indexed: 05/01/2025] Open
Abstract
Pancreatic cancer treatment often relies on multi-drug regimens, but optimal combinations remain elusive. This study evaluates predictive approaches to identify synergistic drug combinations using a dataset from the National Center for Advancing Translational Sciences (NCATS). Screening 496 combinations of 32 anticancer compounds against the PANC-1 cells experimentally determined the degree of synergism and antagonism. Three research groups (NCATS, University of North Carolina, and Massachusetts Institute of Technology) leverage these data to apply machine learning (ML) approaches, predicting synergy across 1.6 million combinations. Of the 88 tested, 51 show synergy, with graph convolutional networks achieving the best hit rate and random forest the highest precision. Beyond highlighting the potential of ML, this work delivers 307 experimentally validated synergistic combinations, demonstrating its practical impact in treating pancreatic cancer.
Collapse
Affiliation(s)
- Mohsen Pourmousa
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Sankalp Jain
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Elena Barnaeva
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Wengong Jin
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Joshua Hochuli
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Zina Itkin
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Travis Maxfield
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Cleber Melo-Filho
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Andrew Thieme
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Kelli Wilson
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Carleen Klumpp-Thomas
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Sam Michael
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Noel Southall
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Tommi Jaakkola
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Eugene N Muratov
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
- Predictive, LLC, Raleigh, NC, 27614, USA
| | - Regina Barzilay
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Alexander Tropsha
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
- Predictive, LLC, Raleigh, NC, 27614, USA
| | - Marc Ferrer
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Alexey V Zakharov
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850, USA.
| |
Collapse
|
2
|
Yu MS, Lee J, Lee Y, Cho D, Oh KS, Jang J, Nong NT, Lee HM, Na D. hERGBoost: A gradient boosting model for quantitative IC 50 prediction of hERG channel blockers. Comput Biol Med 2025; 184:109416. [PMID: 39550914 DOI: 10.1016/j.compbiomed.2024.109416] [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: 06/01/2024] [Revised: 10/25/2024] [Accepted: 11/08/2024] [Indexed: 11/19/2024]
Abstract
The human ether-a-go-go-related gene (hERG) potassium channel is pivotal in drug discovery due to its susceptibility to blockage by drug candidate molecules, which can cause severe cardiotoxic effects. Consequently, identifying and excluding potential hERG channel blockers at the earliest stages of drug development is crucial. Most traditional machine learning models predict a molecule's cardiotoxicity or non-cardiotoxicity typically at 10 μM, which doesn't account for compounds with low IC50 values that are non-toxic at therapeutic levels due to their high effectiveness at lower concentrations. To address the need for more precise, quantitative predictions, we developed hERGBoost, a cutting-edge machine learning model employing a gradient-boosting algorithm. This model demonstrates superior accuracy in predicting the IC50 of drug candidates. Trained on a specially curated dataset for this study, hERGBoost not only exhibited excellent performance in external validation, achieving an R2 score of 0.394 and a low root mean square error of 0.616 but also significantly outstripped previous models in both qualitative and quantitative assessments. Representing a notable leap forward in the prediction of hERG channel blockers, the hERGBoost model and its datasets are freely available to the drug discovery community on our web server at. http://ssbio.cau.ac.kr/software/hergboost This resource promises to be invaluable in advancing safer pharmaceutical development.
Collapse
Affiliation(s)
- Myeong-Sang Yu
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Jingyu Lee
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Yunhyeok Lee
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Daeahn Cho
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Kwang-Seok Oh
- Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea; Department of Medicinal and Pharmaceutical Chemistry, University of Science and Technology, Daejeon, 34129, Republic of Korea
| | - Jidon Jang
- Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea
| | - Nuong Thi Nong
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Hyang-Mi Lee
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Dokyun Na
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Singh D, Alzubi AA, Kaur M, Kumar V, Lee HN. Deep Drug Synergy Prediction Network Using Modified Triangular Mutation-Based Differential Evolution. IEEE J Biomed Health Inform 2025; 29:669-678. [PMID: 38498748 DOI: 10.1109/jbhi.2024.3377631] [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: 03/20/2024]
Abstract
Drug combination therapy is crucial in cancer treatment, but accurately predicting drug synergy remains a challenge due to the complexity of drug combinations. Machine learning and deep learning models have shown promise in drug combination prediction, but they suffer from issues such as gradient vanishing, overfitting, and parameter tuning. To address these problems, the deep drug synergy prediction network, named as EDNet is proposed that leverages a modified triangular mutation-based differential evolution algorithm. This algorithm evolves the initial connection weights and architecture-related attributes of the deep bidirectional mixture density network, improving its performance and addressing the aforementioned issues. EDNet automatically extracts relevant features and provides conditional probability distributions of output attributes. The performance of EDNet is evaluated over two well-known drug synergy datasets, NCI-ALMANAC and deep-synergy. The results demonstrate that EDNet outperforms the competing models. EDNet facilitates efficient drug interactions, enhancing the overall effectiveness of drug combinations for improved cancer treatment outcomes.
Collapse
|
4
|
Ivanov SM, Lagunin AA, Tarasova OA. Analysis of transcription profiles for the identification of master regulators as the key players in glioblastoma. Comput Struct Biotechnol J 2024; 23:3559-3574. [PMID: 39963421 PMCID: PMC11832006 DOI: 10.1016/j.csbj.2024.09.022] [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: 07/08/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 02/20/2025] Open
Abstract
Glioblastoma (GBM) is the most common malignant brain tumor with poor overall survival. Current treatment management for GBM has low efficacy, mainly due to high inter-patient heterogeneity. The transcription profiles in GBM define cell properties essential for tumor progression. We have developed an approach for the identification of master regulators (MRs) that are responsible for the gene expression changes in GBM. The approach is based on transcription factor enrichment analysis with subsequent "upstream" analysis in the signaling network. The main feature of the approach is that all calculations are performed for transcription profiles from individual samples, which allows taking into account GBM transcription heterogeneity. We identified 451 MRs that were up-regulated or down-regulated and, thus, were important parts of positive feedback loops. The number of MRs in the samples correlated with the degree of tumor immune infiltration, while the differences in MR profiles were generally consistent with the known GBM subtypes: mesenchymal, classical, and proneural. MRs densely interact with each other in the signaling network that may be associated with the robustness to pharmacological intervention. We identified 102 receptors among MRs, which is coherent with the importance of cell-cell interactions for GBM progression. The role of some of them in GBM is not currently investigated: lysophosphatidic acid receptors 5 and 6, sphingosine-1-phosphate receptor 4, lysophosphatidylserine receptors GPR34 and GPR174, and G protein-coupled receptors 84 and 132 for fatty acids. Information on the revealed MRs can be used to search for novel therapeutic strategies to treat GBM.
Collapse
Affiliation(s)
- Sergey M. Ivanov
- Department of Bioinformatics, Institute of Biomedical Chemistry, Pogodinskaya Street, 10 bldg. 8, Moscow 119121, Russia
- Department of Bioinformatics, Pirogov Russian National Research Medical University, Ostrovityanova Street, 1, Moscow 117997, Russia
| | - Alexey A. Lagunin
- Department of Bioinformatics, Institute of Biomedical Chemistry, Pogodinskaya Street, 10 bldg. 8, Moscow 119121, Russia
- Department of Bioinformatics, Pirogov Russian National Research Medical University, Ostrovityanova Street, 1, Moscow 117997, Russia
| | - Olga A. Tarasova
- Department of Bioinformatics, Institute of Biomedical Chemistry, Pogodinskaya Street, 10 bldg. 8, Moscow 119121, Russia
| |
Collapse
|
5
|
Ghislat G, Hernandez-Hernandez S, Piyawajanusorn C, Ballester PJ. Data-centric challenges with the application and adoption of artificial intelligence for drug discovery. Expert Opin Drug Discov 2024; 19:1297-1307. [PMID: 39316009 DOI: 10.1080/17460441.2024.2403639] [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: 07/09/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024]
Abstract
INTRODUCTION Artificial intelligence (AI) is exhibiting tremendous potential to reduce the massive costs and long timescales of drug discovery. There are however important challenges currently limiting the impact and scope of AI models. AREAS COVERED In this perspective, the authors discuss a range of data issues (bias, inconsistency, skewness, irrelevance, small size, high dimensionality), how they challenge AI models, and which issue-specific mitigations have been effective. Next, they point out the challenges faced by uncertainty quantification techniques aimed at enhancing and trusting the predictions from these AI models. They also discuss how conceptual errors, unrealistic benchmarks and performance misestimation can confound the evaluation of models and thus their development. Lastly, the authors explain how human bias, whether from AI experts or drug discovery experts, constitutes another challenge that can be alleviated by gaining more prospective experience. EXPERT OPINION AI models are often developed to excel on retrospective benchmarks unlikely to anticipate their prospective performance. As a result, only a few of these models are ever reported to have prospective value (e.g. by discovering potent and innovative drug leads for a therapeutic target). The authors have discussed what can go wrong in practice with AI for drug discovery. The authors hope that this will help inform the decisions of editors, funders investors, and researchers working in this area.
Collapse
Affiliation(s)
- Ghita Ghislat
- Department of Life Sciences, Imperial College London, London, UK
| | | | | | | |
Collapse
|
6
|
Murmu A, Győrffy B. Artificial intelligence methods available for cancer research. Front Med 2024; 18:778-797. [PMID: 39115792 DOI: 10.1007/s11684-024-1085-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/17/2024] [Indexed: 11/01/2024]
Abstract
Cancer is a heterogeneous and multifaceted disease with a significant global footprint. Despite substantial technological advancements for battling cancer, early diagnosis and selection of effective treatment remains a challenge. With the convenience of large-scale datasets including multiple levels of data, new bioinformatic tools are needed to transform this wealth of information into clinically useful decision-support tools. In this field, artificial intelligence (AI) technologies with their highly diverse applications are rapidly gaining ground. Machine learning methods, such as Bayesian networks, support vector machines, decision trees, random forests, gradient boosting, and K-nearest neighbors, including neural network models like deep learning, have proven valuable in predictive, prognostic, and diagnostic studies. Researchers have recently employed large language models to tackle new dimensions of problems. However, leveraging the opportunity to utilize AI in clinical settings will require surpassing significant obstacles-a major issue is the lack of use of the available reporting guidelines obstructing the reproducibility of published studies. In this review, we discuss the applications of AI methods and explore their benefits and limitations. We summarize the available guidelines for AI in healthcare and highlight the potential role and impact of AI models on future directions in cancer research.
Collapse
Affiliation(s)
- Ankita Murmu
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
- National Laboratory for Drug Research and Development, Budapest, 1117, Hungary
- Department of Bioinformatics, Semmelweis University, Budapest, 1094, Hungary
| | - Balázs Győrffy
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary.
- Department of Bioinformatics, Semmelweis University, Budapest, 1094, Hungary.
- Department of Biophysics, University of Pecs, Pecs, 7624, Hungary.
| |
Collapse
|
7
|
Li X, Shen B, Feng F, Li K, Tang Z, Ma L, Li H. Dual-view jointly learning improves personalized drug synergy prediction. Bioinformatics 2024; 40:btae604. [PMID: 39423102 PMCID: PMC11524890 DOI: 10.1093/bioinformatics/btae604] [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: 04/03/2024] [Revised: 08/23/2024] [Accepted: 10/17/2024] [Indexed: 10/21/2024] Open
Abstract
MOTIVATION Accurate and robust estimation of the synergistic drug combination is important for medicine precision. Although some computational methods have been developed, some predictions are still unreliable especially for the cross-dataset predictions, due to the complex mechanism of drug combinations and heterogeneity of cancer samples. RESULTS We have proposed JointSyn that utilizes dual-view jointly learning to predict sample-specific effects of drug combination from drug and cell features. JointSyn outperforms existing state-of-the-art methods in predictive accuracy and robustness across various benchmarks. Each view of JointSyn captures drug synergy-related characteristics and makes complementary contributes to the final prediction of the drug combination. Moreover, JointSyn with fine-tuning improves its generalization ability to predict a novel drug combination or cancer sample using a small number of experimental measurements. We also used JointSyn to generate an estimated atlas of drug synergy for pan-cancer and explored the differential pattern among cancers. These results demonstrate the potential of JointSyn to predict drug synergy, supporting the development of personalized combinatorial therapies. AVAILABILITY AND IMPLEMENTATION Source code and data are available at https://github.com/LiHongCSBLab/JointSyn.
Collapse
Affiliation(s)
- Xueliang Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bihan Shen
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Fangyoumin Feng
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kunshi Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhixuan Tang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Liangxiao Ma
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Science, Shanghai 200031, China
| | - Hong Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
8
|
Nguyen-Vo TH, Do TTT, Nguyen BP. Multitask Learning on Graph Convolutional Residual Neural Networks for Screening of Multitarget Anticancer Compounds. J Chem Inf Model 2024. [PMID: 39197175 DOI: 10.1021/acs.jcim.4c00643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
Recently, various modern experimental screening pipelines and assays have been developed to find promising anticancer drug candidates. However, it is time-consuming and almost infeasible to screen an immense number of compounds for anticancer activity via experimental approaches. To partially address this issue, several computational advances have been proposed. In this study, we present iACP-GCR, a model based on multitask learning on graph convolutional residual neural networks with two types of shortcut connections, to identify multitarget anticancer compounds. In our architecture, the graph convolutional residual neural networks are shared by all the prediction tasks before being separately customized. The NCI-60 data set, one of the most reliable and well-known sources of experimentally verified compounds, was used to develop our model. From that data set, we collected and refined data about compounds screened across nine cancer types (panels), including breast, central nervous system, colon, leukemia, nonsmall cell lung, melanoma, ovarian, prostate, and renal, for model training and evaluation. The model performance evaluated on an independent test set shows that iACP-GCR surpasses the three advanced computational methods for multitask learning. The integration of two shortcut connection types in the shared networks also improves the prediction efficiency. We also deployed the model as a public web server to assist the research community in screening potential anticancer compounds.
Collapse
Affiliation(s)
- Thanh-Hoang Nguyen-Vo
- Ho Chi Minh City Open University, 97 Vo Van Tan, District 3, Ho Chi Minh City 70000, Vietnam
| | - Trang T T Do
- Ho Chi Minh City Open University, 97 Vo Van Tan, District 3, Ho Chi Minh City 70000, Vietnam
| | - Binh P Nguyen
- Victoria University of Wellington, Kelburn Parade, Wellington 6012, New Zealand
| |
Collapse
|
9
|
Zhang H, Chen Y, Payne P, Li F. Using DeepSignalingFlow to mine signaling flows interpreting mechanism of synergy of cocktails. NPJ Syst Biol Appl 2024; 10:92. [PMID: 39169016 PMCID: PMC11339460 DOI: 10.1038/s41540-024-00421-w] [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: 02/13/2024] [Accepted: 08/05/2024] [Indexed: 08/23/2024] Open
Abstract
Complex signaling pathways are believed to be responsible for drug resistance. Drug combinations perturbing multiple signaling targets have the potential to reduce drug resistance. The large-scale multi-omic datasets and experimental drug combination synergistic score data are valuable resources to study mechanisms of synergy (MoS) to guide the development of precision drug combinations. However, signaling patterns of MoS are complex and remain unclear, and thus it is challenging to identify synergistic drug combinations in clinical. Herein, we proposed a novel integrative and interpretable graph AI model, DeepSignalingFlow, to uncover the MoS by integrating and mining multi-omic data. The major innovation is that we uncover MoS by modeling the signaling flow from multi-omic features of essential disease proteins to the drug targets, which has not been introduced by the existing models. The model performance was assessed utilizing four distinct drug combination synergy evaluation datasets, i.e., NCI ALMANAC, O'Neil, DrugComb, and DrugCombDB. The comparison results showed that the proposed model outperformed existing graph AI models in terms of synergy score prediction, and can interpret MoS using the core signaling flows. The code is publicly accessible via Github: https://github.com/FuhaiLiAiLab/DeepSignalingFlow.
Collapse
Affiliation(s)
- Heming Zhang
- Institute for Informatics, Data Science and Biostatistics (I2DB), Washington University School of Medicine, St. Louis, MO, USA
| | - Yixin Chen
- Computer Science, Washington University School of Medicine, St. Louis, MO, USA
| | - Philip Payne
- Institute for Informatics, Data Science and Biostatistics (I2DB), Washington University School of Medicine, St. Louis, MO, USA
| | - Fuhai Li
- Institute for Informatics, Data Science and Biostatistics (I2DB), Washington University School of Medicine, St. Louis, MO, USA.
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
10
|
AlJarf R, Rodrigues CHM, Myung Y, Pires DEV, Ascher DB. piscesCSM: prediction of anticancer synergistic drug combinations. J Cheminform 2024; 16:81. [PMID: 39030592 PMCID: PMC11264925 DOI: 10.1186/s13321-024-00859-4] [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: 08/20/2023] [Accepted: 05/12/2024] [Indexed: 07/21/2024] Open
Abstract
While drug combination therapies are of great importance, particularly in cancer treatment, identifying novel synergistic drug combinations has been a challenging venture. Computational methods have emerged in this context as a promising tool for prioritizing drug combinations for further evaluation, though they have presented limited performance, utility, and interpretability. Here, we propose a novel predictive tool, piscesCSM, that leverages graph-based representations to model small molecule chemical structures to accurately predict drug combinations with favourable anticancer synergistic effects against one or multiple cancer cell lines. Leveraging these insights, we developed a general supervised machine learning model to guide the prediction of anticancer synergistic drug combinations in over 30 cell lines. It achieved an area under the receiver operating characteristic curve (AUROC) of up to 0.89 on independent non-redundant blind tests, outperforming state-of-the-art approaches on both large-scale oncology screening data and an independent test set generated by AstraZeneca (with more than a 16% improvement in predictive accuracy). Moreover, by exploring the interpretability of our approach, we found that simple physicochemical properties and graph-based signatures are predictive of chemotherapy synergism. To provide a simple and integrated platform to rapidly screen potential candidate pairs with favourable synergistic anticancer effects, we made piscesCSM freely available online at https://biosig.lab.uq.edu.au/piscescsm/ as a web server and API. We believe that our predictive tool will provide a valuable resource for optimizing and augmenting combinatorial screening libraries to identify effective and safe synergistic anticancer drug combinations. SCIENTIFIC CONTRIBUTION: This work proposes piscesCSM, a machine-learning-based framework that relies on well-established graph-based representations of small molecules to identify and provide better predictive accuracy of syngenetic drug combinations. Our model, piscesCSM, shows that combining physiochemical properties with graph-based signatures can outperform current architectures on classification prediction tasks. Furthermore, implementing our tool as a web server offers a user-friendly platform for researchers to screen for potential synergistic drug combinations with favorable anticancer effects against one or multiple cancer cell lines.
Collapse
Affiliation(s)
- Raghad AlJarf
- Structural Biology and Bioinformatics, Department of Biochemistry and Pharmacology, University of Melbourne, Melbourne, VIC, Australia
- Systems and Computational Biology, Bio21 Institute, University of Melbourne, Melbourne, VIC, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Carlos H M Rodrigues
- Structural Biology and Bioinformatics, Department of Biochemistry and Pharmacology, University of Melbourne, Melbourne, VIC, Australia
- Systems and Computational Biology, Bio21 Institute, University of Melbourne, Melbourne, VIC, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD, Australia
| | - Yoochan Myung
- Structural Biology and Bioinformatics, Department of Biochemistry and Pharmacology, University of Melbourne, Melbourne, VIC, Australia
- Systems and Computational Biology, Bio21 Institute, University of Melbourne, Melbourne, VIC, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD, Australia
| | - Douglas E V Pires
- Systems and Computational Biology, Bio21 Institute, University of Melbourne, Melbourne, VIC, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- School of Computing and Information Systems, University of Melbourne, Melbourne, VIC, Australia
| | - David B Ascher
- Structural Biology and Bioinformatics, Department of Biochemistry and Pharmacology, University of Melbourne, Melbourne, VIC, Australia.
- Systems and Computational Biology, Bio21 Institute, University of Melbourne, Melbourne, VIC, Australia.
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
11
|
Chen S, Gao N, Li C, Zhai F, Jiang X, Zhang P, Guan J, Li K, Xiang R, Ling G. DrugSK: A Stacked Ensemble Learning Framework for Predicting Drug Combinations of Multiple Diseases. J Chem Inf Model 2024; 64:5317-5327. [PMID: 38900583 DOI: 10.1021/acs.jcim.4c00296] [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: 06/22/2024]
Abstract
Combination therapy is an important direction of continuous exploration in the field of medicine, with the core goals of improving treatment efficacy, reducing adverse reactions, and optimizing clinical outcomes. Machine learning technology holds great promise in improving the prediction of drug synergy combinations. However, most studies focus on single disease-oriented collaborative predictive models or involve excessive feature categories, making it challenging to predict the majority of new drugs. To address these challenges, the DrugSK comprehensive model was developed, which utilizes SMILES-BERT to extract structural information from 3492 drugs and trains on reactions from 48,756 drug combinations. DrugSK is an integrated learning model capable of predicting interactions among various drug categories. First, the primary learner is trained from the initial data set. Random forest, support vector machine, and XGboost model are selected as primary learners and logistic regression as secondary learners. A new data set is then "generated" to train level 2 learners, which can be thought of as a prediction for each model. Finally, the results are filtered using logistic regression. Furthermore, the combination of the new antibacterial drug Drafloxacin with other antibacterial agents was tested. The synergistic effect of Drafloxacin and Isavuconazonium in the fight against Candida albicans has been confirmed, providing enlightenment for the clinical treatment of skin infection. DrugSK's prediction is accurate in practical application and can also predict the probability of the outcome. In addition, the tendency of Drafloxacin and antifungal drugs to be synergistic was found. The development of DrugSK will provide a new blueprint for predicting drug combination synergies.
Collapse
Affiliation(s)
- Siqi Chen
- College of Medical Devices, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Nan Gao
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Chunzhi Li
- College of Medical Devices, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Fei Zhai
- College of Medical Devices, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Xiwei Jiang
- College of Medical Devices, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Peng Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Jibin Guan
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Kefeng Li
- Center for Artificial Intelligence-Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macau SR 999708, China
| | - Rongwu Xiang
- College of Medical Devices, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
- Liaoning Medical Big Data and Artificial Intelligence Engineering Technology Research Center, Shenyang 110016, China
| | - Guixia Ling
- College of Medical Devices, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| |
Collapse
|
12
|
Mao X, Wu S, Huang D, Li C. Complications and comorbidities associated with antineoplastic chemotherapy: Rethinking drug design and delivery for anticancer therapy. Acta Pharm Sin B 2024; 14:2901-2926. [PMID: 39027258 PMCID: PMC11252465 DOI: 10.1016/j.apsb.2024.03.006] [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: 11/26/2023] [Revised: 01/29/2024] [Accepted: 02/10/2024] [Indexed: 07/20/2024] Open
Abstract
Despite the considerable advancements in chemotherapy as a cornerstone modality in cancer treatment, the prevalence of complications and pre-existing diseases is on the rise among cancer patients along with prolonged survival and aging population. The relationships between these disorders and cancer are intricate, bearing significant influence on the survival and quality of life of individuals with cancer and presenting challenges for the prognosis and outcomes of malignancies. Herein, we review the prevailing complications and comorbidities that often accompany chemotherapy and summarize the lessons to learn from inadequate research and management of this scenario, with an emphasis on possible strategies for reducing potential complications and alleviating comorbidities, as well as an overview of current preclinical cancer models and practical advice for establishing bio-faithful preclinical models in such complex context.
Collapse
Affiliation(s)
- Xiaoman Mao
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Shuang Wu
- Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Dandan Huang
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Chong Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
- Medical Research Institute, Southwest University, Chongqing 400715, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| |
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
|
Rafiei F, Zeraati H, Abbasi K, Razzaghi P, Ghasemi JB, Parsaeian M, Masoudi-Nejad A. CFSSynergy: Combining Feature-Based and Similarity-Based Methods for Drug Synergy Prediction. J Chem Inf Model 2024; 64:2577-2585. [PMID: 38514966 DOI: 10.1021/acs.jcim.3c01486] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
Drug synergy prediction plays a vital role in cancer treatment. Because experimental approaches are labor-intensive and expensive, computational-based approaches get more attention. There are two types of computational methods for drug synergy prediction: feature-based and similarity-based. In feature-based methods, the main focus is to extract more discriminative features from drug pairs and cell lines to pass to the task predictor. In similarity-based methods, the similarities among all drugs and cell lines are utilized as features and fed into the task predictor. In this work, a novel approach, called CFSSynergy, that combines these two viewpoints is proposed. First, a discriminative representation is extracted for paired drugs and cell lines as input. We have utilized transformer-based architecture for drugs. For cell lines, we have created a similarity matrix between proteins using the Node2Vec algorithm. Then, the new cell line representation is computed by multiplying the protein-protein similarity matrix and the initial cell line representation. Next, we compute the similarity between unique drugs and unique cells using the learned representation for paired drugs and cell lines. Then, we compute a new representation for paired drugs and cell lines based on the similarity-based features and the learned features. Finally, these features are fed to XGBoost as a task predictor. Two well-known data sets were used to evaluate the performance of our proposed method: DrugCombDB and OncologyScreen. The CFSSynergy approach consistently outperformed existing methods in comparative evaluations. This substantiates the efficacy of our approach in capturing complex synergistic interactions between drugs and cell lines, setting it apart from conventional similarity-based or feature-based methods.
Collapse
Affiliation(s)
- Fatemeh Rafiei
- Department of Epidemiology and Biostatistics, School of Health, Tehran University of Medical Sciences, Tehran 14167-53955, Iran
| | - Hojjat Zeraati
- Department of Epidemiology and Biostatistics, School of Health, Tehran University of Medical Sciences, Tehran 14167-53955, Iran
| | - Karim Abbasi
- Laboratory of System Biology, Bioinformatics & Artificial Intelligence in Medicine (LBB&AI), Faculty of Mathematics and Computer Science, Kharazmi University, Tehran 14588-89694, Iran
| | - Parvin Razzaghi
- Department of Computer Science and Information Technology, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 45137-66731, Iran
| | - Jahan B Ghasemi
- Chemistry Department, Faculty of Chemistry, School of Sciences, University of Tehran, Tehran 14174-66191, Iran
| | - Mahboubeh Parsaeian
- Department of Epidemiology and Biostatistics, School of Health, Tehran University of Medical Sciences, Tehran 14167-53955, Iran
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, U.K
| | - Ali Masoudi-Nejad
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran 13145-1365, Iran
| |
Collapse
|
15
|
Alam W, Tayara H, Chong KT. Unlocking the therapeutic potential of drug combinations through synergy prediction using graph transformer networks. Comput Biol Med 2024; 170:108007. [PMID: 38242015 DOI: 10.1016/j.compbiomed.2024.108007] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 01/03/2024] [Accepted: 01/13/2024] [Indexed: 01/21/2024]
Abstract
Drug combinations are frequently used to treat cancer to reduce side effects and increase efficacy. The experimental discovery of drug combination synergy is time-consuming and expensive for large datasets. Therefore, an efficient and reliable computational approach is required to investigate these drug combinations. Advancements in deep learning can handle large datasets with various biological problems. In this study, we developed a SynergyGTN model based on the Graph Transformer Network to predict the synergistic drug combinations against an untreated cancer cell line expression profile. We represent the drug via a graph, with each node and edge of the graph containing nine types of atomic feature vectors and four bonds features, respectively. The cell lines represent based on their gene expression profiles. The drug graph was passed through the GTN layers to extract a generalized feature map for each drug pairs. The drug pair extracted features and cell-line gene expression profiles were concatenated and subsequently subjected to processing through multiple densely connected layers. SynergyGTN outperformed the state-of-the-art methods, with a receiver operating characteristic area under the curve improvement of 5% on the 5-fold cross-validation. The accuracy of SynergyGTN was further verified through three types of cross-validation tests strategies namely leave-drug-out, leave-combination-out, and leave-tissue-out, resulting in improvement in accuracy of 8%, 1%, and 2%, respectively. The Astrazeneca Dream dataset was utilized as an independent dataset to validate and assess the generalizability of the proposed method, resulting in an improvement in balanced accuracy of 13%. In conclusion, SynergyGTN is a reliable and efficient computational approach for predicting drug combination synergy in cancer treatment. Finally, we developed a web server tool to facilitate the pharmaceutical industry and researchers, as available at: http://nsclbio.jbnu.ac.kr/tools/SynergyGTN/.
Collapse
Affiliation(s)
- Waleed Alam
- Department of Electronics and Information Engineering, Jeonbuk National University, Jeonju, 54896, South Korea
| | - Hilal Tayara
- School of International Engineering and Science, Jeonbuk National University, Jeonju, 54896, South Korea.
| | - Kil To Chong
- Department of Electronics and Information Engineering, Jeonbuk National University, Jeonju, 54896, South Korea; Advanced Electronics and Information Research Center, Jeonbuk National University, Jeonju, 54896, South Korea.
| |
Collapse
|
16
|
Taoma K, Ruengjitchatchawalya M, Liangruksa M, Laomettachit T. Boolean modeling of breast cancer signaling pathways uncovers mechanisms of drug synergy. PLoS One 2024; 19:e0298788. [PMID: 38394152 PMCID: PMC10889607 DOI: 10.1371/journal.pone.0298788] [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: 09/25/2023] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Breast cancer is one of the most common types of cancer in females. While drug combinations have shown potential in breast cancer treatments, identifying new effective drug pairs is challenging due to the vast number of possible combinations among available compounds. Efforts have been made to accelerate the process with in silico predictions. Here, we developed a Boolean model of signaling pathways in breast cancer. The model was tailored to represent five breast cancer cell lines by integrating information about cell-line specific mutations, gene expression, and drug treatments. The models reproduced cell-line specific protein activities and drug-response behaviors in agreement with experimental data. Next, we proposed a calculation of protein synergy scores (PSSs), determining the effect of drug combinations on individual proteins' activities. The PSSs of selected proteins were used to investigate the synergistic effects of 150 drug combinations across five cancer cell lines. The comparison of the highest single agent (HSA) synergy scores between experiments and model predictions from the MDA-MB-231 cell line achieved the highest Pearson's correlation coefficient of 0.58 with a great balance among the classification metrics (AUC = 0.74, sensitivity = 0.63, and specificity = 0.64). Finally, we clustered drug pairs into groups based on the selected PSSs to gain further insights into the mechanisms underlying the observed synergistic effects of drug pairs. Clustering analysis allowed us to identify distinct patterns in the protein activities that correspond to five different modes of synergy: 1) synergistic activation of FADD and BID (extrinsic apoptosis pathway), 2) synergistic inhibition of BCL2 (intrinsic apoptosis pathway), 3) synergistic inhibition of MTORC1, 4) synergistic inhibition of ESR1, and 5) synergistic inhibition of CYCLIN D. Our approach offers a mechanistic understanding of the efficacy of drug combinations and provides direction for selecting potential drug pairs worthy of further laboratory investigation.
Collapse
Affiliation(s)
- Kittisak Taoma
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology, King Mongkut’s University of Technology Thonburi, Bangkok, Thailand
- School of Information Technology, King Mongkut’s University of Technology Thonburi, Bangkok, Thailand
| | - Marasri Ruengjitchatchawalya
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology, King Mongkut’s University of Technology Thonburi, Bangkok, Thailand
- Biotechnology Program, School of Bioresources and Technology, King Mongkut’s University of Technology Thonburi, Bangkok, Thailand
| | - Monrudee Liangruksa
- National Nanotechnology Center, National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Teeraphan Laomettachit
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology, King Mongkut’s University of Technology Thonburi, Bangkok, Thailand
- Theoretical and Computational Physics Group, Center of Excellence in Theoretical and Computational Science, King Mongkut’s University of Technology Thonburi, Bangkok, Thailand
| |
Collapse
|
17
|
Li T, Shetty S, Kamath A, Jaiswal A, Jiang X, Ding Y, Kim Y. CancerGPT for few shot drug pair synergy prediction using large pretrained language models. NPJ Digit Med 2024; 7:40. [PMID: 38374445 PMCID: PMC10876664 DOI: 10.1038/s41746-024-01024-9] [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: 05/19/2023] [Accepted: 02/02/2024] [Indexed: 02/21/2024] Open
Abstract
Large language models (LLMs) have been shown to have significant potential in few-shot learning across various fields, even with minimal training data. However, their ability to generalize to unseen tasks in more complex fields, such as biology and medicine has yet to be fully evaluated. LLMs can offer a promising alternative approach for biological inference, particularly in cases where structured data and sample size are limited, by extracting prior knowledge from text corpora. Here we report our proposed few-shot learning approach, which uses LLMs to predict the synergy of drug pairs in rare tissues that lack structured data and features. Our experiments, which involved seven rare tissues from different cancer types, demonstrate that the LLM-based prediction model achieves significant accuracy with very few or zero samples. Our proposed model, the CancerGPT (with ~ 124M parameters), is comparable to the larger fine-tuned GPT-3 model (with ~ 175B parameters). Our research contributes to tackling drug pair synergy prediction in rare tissues with limited data, and also advancing the use of LLMs for biological and medical inference tasks.
Collapse
Affiliation(s)
- Tianhao Li
- School of Information, University of Texas at Austin, Austin, TX, USA
| | - Sandesh Shetty
- Manning College of Information and Computer Sciences, University of Massachusetts Amherst, Amherst, MA, USA
| | - Advaith Kamath
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Ajay Jaiswal
- School of Information, University of Texas at Austin, Austin, TX, USA
| | - Xiaoqian Jiang
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ying Ding
- School of Information, University of Texas at Austin, Austin, TX, USA
| | - Yejin Kim
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
18
|
Abd El-Hafeez T, Shams MY, Elshaier YAMM, Farghaly HM, Hassanien AE. Harnessing machine learning to find synergistic combinations for FDA-approved cancer drugs. Sci Rep 2024; 14:2428. [PMID: 38287066 PMCID: PMC10825182 DOI: 10.1038/s41598-024-52814-w] [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: 11/09/2023] [Accepted: 01/24/2024] [Indexed: 01/31/2024] Open
Abstract
Combination therapy is a fundamental strategy in cancer chemotherapy. It involves administering two or more anti-cancer agents to increase efficacy and overcome multidrug resistance compared to monotherapy. However, drug combinations can exhibit synergy, additivity, or antagonism. This study presents a machine learning framework to classify and predict cancer drug combinations. The framework utilizes several key steps including data collection and annotation from the O'Neil drug interaction dataset, data preprocessing, stratified splitting into training and test sets, construction and evaluation of classification models to categorize combinations as synergistic, additive, or antagonistic, application of regression models to predict combination sensitivity scores for enhanced predictions compared to prior work, and the last step is examination of drug features and mechanisms of action to understand synergy behaviors for optimal combinations. The models identified combination pairs most likely to synergize against different cancers. Kinase inhibitors combined with mTOR inhibitors, DNA damage-inducing drugs or HDAC inhibitors showed benefit, particularly for ovarian, melanoma, prostate, lung and colorectal carcinomas. Analysis highlighted Gemcitabine, MK-8776 and AZD1775 as frequently synergizing across cancer types. This machine learning framework provides a valuable approach to uncover more effective multi-drug regimens.
Collapse
Affiliation(s)
- Tarek Abd El-Hafeez
- Department of Computer Science, Faculty of Science, Minia University, El-Minia, Egypt.
- Computer Science Unit, Deraya University, El-Minia, Egypt.
| | - Mahmoud Y Shams
- Faculty of Artificial Intelligence, Kafrelsheikh University, Kafr El-Sheikh, Egypt
- Scientific Research Group in Egypt (SRGE), Cairo, Egypt
| | - Yaseen A M M Elshaier
- Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, Menoufia, Egypt
| | - Heba Mamdouh Farghaly
- Department of Computer Science, Faculty of Science, Minia University, El-Minia, Egypt
| | - Aboul Ella Hassanien
- Faculty of Computers and Artificial Intelligence, Cairo University, Cairo, Egypt.
- Scientific Research Group in Egypt (SRGE), Cairo, Egypt.
| |
Collapse
|
19
|
Liu M, Srivastava G, Ramanujam J, Brylinski M. Augmented drug combination dataset to improve the performance of machine learning models predicting synergistic anticancer effects. Sci Rep 2024; 14:1668. [PMID: 38238448 PMCID: PMC10796434 DOI: 10.1038/s41598-024-51940-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/11/2024] [Indexed: 01/22/2024] Open
Abstract
Combination therapy has gained popularity in cancer treatment as it enhances the treatment efficacy and overcomes drug resistance. Although machine learning (ML) techniques have become an indispensable tool for discovering new drug combinations, the data on drug combination therapy currently available may be insufficient to build high-precision models. We developed a data augmentation protocol to unbiasedly scale up the existing anti-cancer drug synergy dataset. Using a new drug similarity metric, we augmented the synergy data by substituting a compound in a drug combination instance with another molecule that exhibits highly similar pharmacological effects. Using this protocol, we were able to upscale the AZ-DREAM Challenges dataset from 8798 to 6,016,697 drug combinations. Comprehensive performance evaluations show that ML models trained on the augmented data consistently achieve higher accuracy than those trained solely on the original dataset. Our data augmentation protocol provides a systematic and unbiased approach to generating more diverse and larger-scale drug combination datasets, enabling the development of more precise and effective ML models. The protocol presented in this study could serve as a foundation for future research aimed at discovering novel and effective drug combinations for cancer treatment.
Collapse
Affiliation(s)
- Mengmeng Liu
- Division of Electrical and Computer Engineering, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - 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
- 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
|
20
|
Rani P, Dutta K, Kumar V. Performance evaluation of drug synergy datasets using computational intelligence approaches. MULTIMEDIA TOOLS AND APPLICATIONS 2024; 83:8971-8997. [DOI: 10.1007/s11042-023-15723-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/26/2022] [Accepted: 04/18/2023] [Indexed: 01/03/2025]
|
21
|
Guendouzi A, Belkhiri L, Guendouzi A, Derouiche TMT, Djekoun A. A combined in silico approaches of 2D-QSAR, molecular docking, molecular dynamics and ADMET prediction of anti-cancer inhibitor activity for actinonin derivatives. J Biomol Struct Dyn 2024; 42:119-133. [PMID: 36995063 DOI: 10.1080/07391102.2023.2192801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/10/2023] [Indexed: 03/31/2023]
Abstract
Inhibition of human mitochondrial peptide deformylase (HsPDF) plays a major role in reducing growth, proliferation, and cellular cancer survival. In this work, a series of 32 actinonin derivatives for HsPDF (PDB: 3G5K) inhibitor's anticancer activity was computationally analyzed for the first time, using an in silico study considering 2D-QSAR modeling, and molecular docking studies, and validated by molecular dynamics and ADMET properties. The results of multilinear regression (MLR) and artificial neural networks (ANN) statistical analysis reveal a good correlation between pIC50 activity and the seven (7) descriptors. The developed models were highly significant with cross-validation, the Y-randomization test and their applicability range. In addition, all considered data sets show that the AC30 compound, exhibits the best binding affinity (docking score = -212.074 kcal/mol and H-bonding energy = -15.879 kcal/mol). Furthermore, molecular dynamics simulations were performed at 500 ns, confirming the stability of the studied complexes under physiological conditions and validating the molecular docking results. Five selected actinonin derivatives (AC1, AC8, AC15, AC18 and AC30), exhibiting best docking score, were rationalized as potential leads for HsPDF inhibition, in well agreement with experimental outcomes. Furthermore, based on the in silico study, new six molecules (AC32, AC33, AC34, AC35, AC36 and AC37) were suggested as HsPDF inhibition candidates, which would be combined with in-vitro and in-vivo studies to perspective validation of their anticancer activity. Indeed, the ADMET predictions indicate that these six new ligands have demonstrated a fairly good drug-likeness profile.
Collapse
Affiliation(s)
| | - Lotfi Belkhiri
- Centre de Recherche en Sciences Pharmaceutiques CRSP, Constantine, Algeria
- Laboratoire de Physique Mathématique et Subatomique LPMS, Département de Chimie, Université des Frères Mentouri, Constantine, Algeria
| | - Abdelkrim Guendouzi
- Laboratoire de Chimie, Synthèse, Propriétés et Applications LCSPA, Département de Chimie, Faculté des Sciences, Université Dr Moulay Tahar de Saida, Saïda, Algeria
| | - Tahar Mohamed Taha Derouiche
- Centre de Recherche en Sciences Pharmaceutiques CRSP, Constantine, Algeria
- Laboratoire Innovation Développement des Actifs Pharmaceutiques LIDAP, Faculté de Médecine, Département Pharmacie, Université Salah Boubnider Constantine 3, El Khroub, Algeria
| | - Abdelhamid Djekoun
- Centre de Recherche en Sciences Pharmaceutiques CRSP, Constantine, Algeria
| |
Collapse
|
22
|
Yan K, Jia R, Guo S. SynAI: an AI-driven cancer drugs synergism prediction platform. BIOINFORMATICS ADVANCES 2023; 3:vbad160. [PMID: 38023331 PMCID: PMC10660295 DOI: 10.1093/bioadv/vbad160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/13/2023] [Accepted: 11/08/2023] [Indexed: 12/01/2023]
Abstract
Summary The SynAI solution is a flexible AI-driven drug synergism prediction solution aiming to discover potential therapeutic value of compounds in early stage. Rather than providing a finite choice of drug combination or cell lines, SynAI is capable of predicting potential drug synergism/antagonism using in silico compound SMILE (Simplified Molecular Input Line Entry System) sequences. The AI core of SynAI platform has been trained against cell lines and compound pairs listed by NCI (National Cancer Institute)-Almanac and DurgCombDB datasets. In total, the training data consists of over 1 200 000 in vitro synergism tests on 150 cancer cell lines of different organ origins. Each cell line is tested against over 6000 pairs of FDA (Food and Drug Administration) approved compound combinations. Given one or both candidate compound in SMILE sequence, SynAI is able to predict the potential Bliss score of the combined compound test with the designated cell line without the needs of compound synthetization or structural analysis; thus can significantly reduce the candidate screening costs during the compound development. SynAI platform demonstrates a comparable performance to existing methods but offers more flexibilities for data input. Availability and implementation The evaluation version of SynAI is freely accessible online at https://synai.crownbio.com.
Collapse
Affiliation(s)
- Kuan Yan
- Data Science and Bioinformatics, Crown Bioscience, Suzhou, Jiangsu Province 215000, P.R. China
| | - Runjun Jia
- Data Science and Bioinformatics, Crown Bioscience, Suzhou, Jiangsu Province 215000, P.R. China
| | - Sheng Guo
- Data Science and Bioinformatics, Crown Bioscience, Suzhou, Jiangsu Province 215000, P.R. China
| |
Collapse
|
23
|
Liu M, Srivastava G, Ramanujam J, Brylinski M. Augmented drug combination dataset to improve the performance of machine learning models predicting synergistic anticancer effects. RESEARCH SQUARE 2023:rs.3.rs-3481858. [PMID: 37961281 PMCID: PMC10635365 DOI: 10.21203/rs.3.rs-3481858/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Combination therapy has gained popularity in cancer treatment as it enhances the treatment efficacy and overcomes drug resistance. Although machine learning (ML) techniques have become an indispensable tool for discovering new drug combinations, the data on drug combination therapy currently available may be insufficient to build high-precision models. We developed a data augmentation protocol to unbiasedly scale up the existing anti-cancer drug synergy dataset. Using a new drug similarity metric, we augmented the synergy data by substituting a compound in a drug combination instance with another molecule that exhibits highly similar pharmacological effects. Using this protocol, we were able to upscale the AZ-DREAM Challenges dataset from 8,798 to 6,016,697 drug combinations. Comprehensive performance evaluations show that Random Forest and Gradient Boosting Trees models trained on the augmented data achieve higher accuracy than those trained solely on the original dataset. Our data augmentation protocol provides a systematic and unbiased approach to generating more diverse and larger-scale drug combination datasets, enabling the development of more precise and effective ML models. The protocol presented in this study could serve as a foundation for future research aimed at discovering novel and effective drug combinations for cancer treatment.
Collapse
|
24
|
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
|
25
|
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
|
26
|
Pan Y, Ren H, Lan L, Li Y, Huang T. Review of Predicting Synergistic Drug Combinations. Life (Basel) 2023; 13:1878. [PMID: 37763281 PMCID: PMC10533134 DOI: 10.3390/life13091878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
The prediction of drug combinations is of great clinical significance. In many diseases, such as high blood pressure, diabetes, and stomach ulcers, the simultaneous use of two or more drugs has shown clear efficacy. It has greatly reduced the progression of drug resistance. This review presents the latest applications of methods for predicting the effects of drug combinations and the bioactivity databases commonly used in drug combination prediction. These studies have played a significant role in developing precision therapy. We first describe the concept of synergy. we study various publicly available databases for drug combination prediction tasks. Next, we introduce five algorithms applied to drug combinatorial prediction, which include traditional machine learning methods, deep learning methods, mathematical methods, systems biology methods and search algorithms. In the end, we sum up the difficulties encountered in prediction models.
Collapse
Affiliation(s)
- Yichen Pan
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; (Y.P.); (H.R.)
| | - Haotian Ren
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; (Y.P.); (H.R.)
| | - Liang Lan
- Department of Interactive Media, Hong Kong Baptist University, Hong Kong, China;
| | - Yixue Li
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; (Y.P.); (H.R.)
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- Guangzhou Laboratory, Guangzhou 510005, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai 200433, China
| | - Tao Huang
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; (Y.P.); (H.R.)
| |
Collapse
|
27
|
Dong Z, Zhang H, Chen Y, Payne PRO, Li F. Interpreting the Mechanism of Synergism for Drug Combinations Using Attention-Based Hierarchical Graph Pooling. Cancers (Basel) 2023; 15:4210. [PMID: 37686486 PMCID: PMC10486573 DOI: 10.3390/cancers15174210] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/16/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
Synergistic drug combinations provide huge potentials to enhance therapeutic efficacy and to reduce adverse reactions. However, effective and synergistic drug combination prediction remains an open question because of the unknown causal disease signaling pathways. Though various deep learning (AI) models have been proposed to quantitatively predict the synergism of drug combinations, the major limitation of existing deep learning methods is that they are inherently not interpretable, which makes the conclusions of AI models untransparent to human experts, henceforth limiting the robustness of the model conclusion and the implementation ability of these models in real-world human-AI healthcare. In this paper, we develop an interpretable graph neural network (GNN) that reveals the underlying essential therapeutic targets and the mechanism of the synergy (MoS) by mining the sub-molecular network of great importance. The key point of the interpretable GNN prediction model is a novel graph pooling layer, a self-attention-based node and edge pool (henceforth SANEpool), that can compute the attention score (importance) of genes and connections based on the genomic features and topology. As such, the proposed GNN model provides a systematic way to predict and interpret the drug combination synergism based on the detected crucial sub-molecular network. Experiments on various well-adopted drug-synergy-prediction datasets demonstrate that (1) the SANEpool model has superior predictive ability to generate accurate synergy score prediction, and (2) the sub-molecular networks detected by the SANEpool are self-explainable and salient for identifying synergistic drug combinations.
Collapse
Affiliation(s)
- Zehao Dong
- Department of Computer Science & Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA; (Z.D.); (Y.C.)
| | - Heming Zhang
- Institute for Informatics, Data Science, and Biostatistics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; (H.Z.); (P.R.O.P.)
| | - Yixin Chen
- Department of Computer Science & Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA; (Z.D.); (Y.C.)
| | - Philip R. O. Payne
- Institute for Informatics, Data Science, and Biostatistics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; (H.Z.); (P.R.O.P.)
| | - Fuhai Li
- Institute for Informatics, Data Science, and Biostatistics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; (H.Z.); (P.R.O.P.)
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
28
|
Rafiei F, Zeraati H, Abbasi K, Ghasemi JB, Parsaeian M, Masoudi-Nejad A. DeepTraSynergy: drug combinations using multimodal deep learning with transformers. Bioinformatics 2023; 39:btad438. [PMID: 37467066 PMCID: PMC10397534 DOI: 10.1093/bioinformatics/btad438] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 06/27/2023] [Accepted: 07/17/2023] [Indexed: 07/21/2023] Open
Abstract
MOTIVATION Screening bioactive compounds in cancer cell lines receive more attention. Multidisciplinary drugs or drug combinations have a more effective role in treatments and selectively inhibit the growth of cancer cells. RESULTS Hence, we propose a new deep learning-based approach for drug combination synergy prediction called DeepTraSynergy. Our proposed approach utilizes multimodal input including drug-target interaction, protein-protein interaction, and cell-target interaction to predict drug combination synergy. To learn the feature representation of drugs, we have utilized transformers. It is worth noting that our approach is a multitask approach that predicts three outputs including the drug-target interaction, its toxic effect, and drug combination synergy. In our approach, drug combination synergy is the main task and the two other ones are the auxiliary tasks that help the approach to learn a better model. In the proposed approach three loss functions are defined: synergy loss, toxic loss, and drug-protein interaction loss. The last two loss functions are designed as auxiliary losses to help learn a better solution. DeepTraSynergy outperforms the classic and state-of-the-art models in predicting synergistic drug combinations on the two latest drug combination datasets. The DeepTraSynergy algorithm achieves accuracy values of 0.7715 and 0.8052 (an improvement over other approaches) on the DrugCombDB and Oncology-Screen datasets, respectively. Also, we evaluate the contribution of each component of DeepTraSynergy to show its effectiveness in the proposed method. The introduction of the relation between proteins (PPI networks) and drug-protein interaction significantly improves the prediction of synergistic drug combinations. AVAILABILITY AND IMPLEMENTATION The source code and data are available at https://github.com/fatemeh-rafiei/DeepTraSynergy.
Collapse
Affiliation(s)
- Fatemeh Rafiei
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran 1417613151, Iran
| | - Hojjat Zeraati
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran 1417613151, Iran
| | - Karim Abbasi
- Laboratory of System Biology, Bioinformatics & Artificial Intelligent in Medicine (LBB&AI), Faculty of Mathematics and Computer Science, Kharazmi University, Tehran 1571914911, Iran
| | - Jahan B Ghasemi
- Chemistry Department, Faculty of Chemistry, School of Sciences, University of Tehran, Tehran 1417614411, Iran
| | - Mahboubeh Parsaeian
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran 1417613151, Iran
- Department of Epidemiology & Biostatistics, School of Public Health, Imperial College London, London W21PG, United Kingdom
| | - Ali Masoudi-Nejad
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran 1417614411, Iran
| |
Collapse
|
29
|
Hariri A, Mirian M, Zarrabi A, Kohandel M, Amini-Pozveh M, Aref AR, Tabatabaee A, Prabhakar PK, Sivakumar PM. The circadian rhythm: an influential soundtrack in the diabetes story. Front Endocrinol (Lausanne) 2023; 14:1156757. [PMID: 37441501 PMCID: PMC10333930 DOI: 10.3389/fendo.2023.1156757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/03/2023] [Indexed: 07/15/2023] Open
Abstract
Type 2 Diabetes Mellitus (T2DM) has been the main category of metabolic diseases in recent years due to changes in lifestyle and environmental conditions such as diet and physical activity. On the other hand, the circadian rhythm is one of the most significant biological pathways in humans and other mammals, which is affected by light, sleep, and human activity. However, this cycle is controlled via complicated cellular pathways with feedback loops. It is widely known that changes in the circadian rhythm can alter some metabolic pathways of body cells and could affect the treatment process, particularly for metabolic diseases like T2DM. The aim of this study is to explore the importance of the circadian rhythm in the occurrence of T2DM via reviewing the metabolic pathways involved, their relationship with the circadian rhythm from two perspectives, lifestyle and molecular pathways, and their effect on T2DM pathophysiology. These impacts have been demonstrated in a variety of studies and led to the development of approaches such as time-restricted feeding, chronotherapy (time-specific therapies), and circadian molecule stabilizers.
Collapse
Affiliation(s)
- Amirali Hariri
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mina Mirian
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, Türkiye
| | - Mohammad Kohandel
- Department of Applied Mathematics, Faculty of Mathematics, University of Waterloo, Waterloo, ON, Canada
| | - Maryam Amini-Pozveh
- Department of Prosthodontics Dentistry, Dental Materials Research Center, Dental Research Institute, School of Dentistry, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana Farber Cancer Institute, Boston, MA, United States
- Translational Sciences, Xsphera Biosciences Inc., Boston, MA, United States
| | - Aliye Tabatabaee
- School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Pranav Kumar Prabhakar
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Lovely Professional University, Phagwara, Punjab, India
- Division of Research and Development, Lovely Professional University, Phagwara Punjab, India
| | - Ponnurengam Malliappan Sivakumar
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam
- School of Medicine and Pharmacy, Duy Tan University, Da Nang, Vietnam
| |
Collapse
|
30
|
Li T, Shetty S, Kamath A, Jaiswal A, Jiang X, Ding Y, Kim Y. CancerGPT: Few-shot Drug Pair Synergy Prediction using Large Pre-trained Language Models. ARXIV 2023:arXiv:2304.10946v1. [PMID: 37131872 PMCID: PMC10153348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Large pre-trained language models (LLMs) have been shown to have significant potential in few-shot learning across various fields, even with minimal training data. However, their ability to generalize to unseen tasks in more complex fields, such as biology, has yet to be fully evaluated. LLMs can offer a promising alternative approach for biological inference, particularly in cases where structured data and sample size are limited, by extracting prior knowledge from text corpora. Our proposed few-shot learning approach uses LLMs to predict the synergy of drug pairs in rare tissues that lack structured data and features. Our experiments, which involved seven rare tissues from different cancer types, demonstrated that the LLM-based prediction model achieved significant accuracy with very few or zero samples. Our proposed model, the CancerGPT (with ~ 124M parameters), was even comparable to the larger fine-tuned GPT-3 model (with ~ 175B parameters). Our research is the first to tackle drug pair synergy prediction in rare tissues with limited data. We are also the first to utilize an LLM-based prediction model for biological reaction prediction tasks.
Collapse
Affiliation(s)
- Tianhao Li
- School of Information, University of Texas at Austin, Austin, Texas, USA
| | - Sandesh Shetty
- College of Information and Computer Sciences, University of Massachusetts Amherst, Amherst, MA, USA
| | - Advaith Kamath
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Ajay Jaiswal
- School of Information, University of Texas at Austin, Austin, TX, USA
| | - Xiaoqian Jiang
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ying Ding
- School of Information, University of Texas at Austin, Austin, TX, USA
| | - Yejin Kim
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
31
|
Torkamannia A, Omidi Y, Ferdousi R. SYNDEEP: a deep learning approach for the prediction of cancer drugs synergy. Sci Rep 2023; 13:6184. [PMID: 37061563 PMCID: PMC10105711 DOI: 10.1038/s41598-023-33271-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 04/11/2023] [Indexed: 04/17/2023] Open
Abstract
Drug combinations can be the prime strategy for increasing the initial treatment options in cancer therapy. However, identifying the combinations through experimental approaches is very laborious and costly. Notably, in vitro and/or in vivo examination of all the possible combinations might not be plausible. This study presented a novel computational approach to predicting synergistic drug combinations. Specifically, the deep neural network-based binary classification was utilized to develop the model. Various physicochemical, genomic, protein-protein interaction and protein-metabolite interaction information were used to predict the synergy effects of the combinations of different drugs. The performance of the constructed model was compared with shallow neural network (SNN), k-nearest neighbors (KNN), random forest (RF), support vector machines (SVMs), and gradient boosting classifiers (GBC). Based on our findings, the proposed deep neural network model was found to be capable of predicting synergistic drug combinations with high accuracy. The prediction accuracy and AUC metrics for this model were 92.21% and 97.32% in tenfold cross-validation. According to the results, the integration of different types of physicochemical and genomics features leads to more accurate prediction of synergy in cancer drugs.
Collapse
Affiliation(s)
- Anna Torkamannia
- Department of Health Information Technology, School of Management and Medical Informatics, Tabriz University of Medical Sciences, Tabriz, 51656/65811, Iran
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, 33328, USA
| | - Reza Ferdousi
- Department of Health Information Technology, School of Management and Medical Informatics, Tabriz University of Medical Sciences, Tabriz, 51656/65811, Iran.
| |
Collapse
|
32
|
Zhang H, Wang Z, Nan Y, Zagidullin B, Yi D, Tang J, Guan Y. Harmonizing across datasets to improve the transferability of drug combination prediction. Commun Biol 2023; 6:397. [PMID: 37041243 PMCID: PMC10090076 DOI: 10.1038/s42003-023-04783-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/30/2023] [Indexed: 04/13/2023] Open
Abstract
Combination treatment has multiple advantages over traditional monotherapy in clinics, thus becoming a target of interest for many high-throughput screening (HTS) studies, which enables the development of machine learning models predicting the response of new drug combinations. However, most existing models have been tested only within a single study, and these models cannot generalize across different datasets due to significantly variable experimental settings. Here, we thoroughly assessed the transferability issue of single-study-derived models on new datasets. More importantly, we propose a method to overcome the experimental variability by harmonizing dose-response curves of different studies. Our method improves the prediction performance of machine learning models by 184% and 1367% compared to the baseline models in intra-study and inter-study predictions, respectively, and shows consistent improvement in multiple cross-validation settings. Our study addresses the crucial question of the transferability in drug combination predictions, which is fundamental for such models to be extrapolated to new drug combination discovery and clinical applications that are de facto different datasets.
Collapse
Affiliation(s)
- Hanrui Zhang
- Department of Computational Medicine and Bioinformatics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Ziyan Wang
- Department of Electrical Engineering and Computer Science (EECS) - CSE Division, University of Michigan, Ann Arbor, MI, USA
| | - Yiyang Nan
- Department of Computational Medicine and Bioinformatics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Bulat Zagidullin
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Daiyao Yi
- Department of Computational Medicine and Bioinformatics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Jing Tang
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Yuanfang Guan
- Department of Computational Medicine and Bioinformatics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA.
- Department of Internal medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
33
|
Baptista D, Ferreira PG, Rocha M. A systematic evaluation of deep learning methods for the prediction of drug synergy in cancer. PLoS Comput Biol 2023; 19:e1010200. [PMID: 36952569 PMCID: PMC10072473 DOI: 10.1371/journal.pcbi.1010200] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 04/04/2023] [Accepted: 02/08/2023] [Indexed: 03/25/2023] Open
Abstract
One of the main obstacles to the successful treatment of cancer is the phenomenon of drug resistance. A common strategy to overcome resistance is the use of combination therapies. However, the space of possibilities is huge and efficient search strategies are required. Machine Learning (ML) can be a useful tool for the discovery of novel, clinically relevant anti-cancer drug combinations. In particular, deep learning (DL) has become a popular choice for modeling drug combination effects. Here, we set out to examine the impact of different methodological choices on the performance of multimodal DL-based drug synergy prediction methods, including the use of different input data types, preprocessing steps and model architectures. Focusing on the NCI ALMANAC dataset, we found that feature selection based on prior biological knowledge has a positive impact-limiting gene expression data to cancer or drug response-specific genes improved performance. Drug features appeared to be more predictive of drug response, with a 41% increase in coefficient of determination (R2) and 26% increase in Spearman correlation relative to a baseline model that used only cell line and drug identifiers. Molecular fingerprint-based drug representations performed slightly better than learned representations-ECFP4 fingerprints increased R2 by 5.3% and Spearman correlation by 2.8% w.r.t the best learned representations. In general, fully connected feature-encoding subnetworks outperformed other architectures. DL outperformed other ML methods by more than 35% (R2) and 14% (Spearman). Additionally, an ensemble combining the top DL and ML models improved performance by about 6.5% (R2) and 4% (Spearman). Using a state-of-the-art interpretability method, we showed that DL models can learn to associate drug and cell line features with drug response in a biologically meaningful way. The strategies explored in this study will help to improve the development of computational methods for the rational design of effective drug combinations for cancer therapy.
Collapse
Affiliation(s)
- Delora Baptista
- CEB - Centre of Biological Engineering, University of Minho, Braga, Portugal
- LABBELS - Associate Laboratory, Braga, Guimarães, Portugal
| | - Pedro G Ferreira
- Department of Computer Science, Faculty of Sciences, University of Porto, Porto, Portugal
- INESC TEC, Porto, Portugal
- Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- i3s - Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal
| | - Miguel Rocha
- CEB - Centre of Biological Engineering, University of Minho, Braga, Portugal
- LABBELS - Associate Laboratory, Braga, Guimarães, Portugal
| |
Collapse
|
34
|
Xu M, Zhao X, Wang J, Feng W, Wen N, Wang C, Wang J, Liu Y, Zhao L. DFFNDDS: prediction of synergistic drug combinations with dual feature fusion networks. J Cheminform 2023; 15:33. [PMID: 36927504 PMCID: PMC10022091 DOI: 10.1186/s13321-023-00690-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/30/2023] [Indexed: 03/18/2023] Open
Abstract
Drug combination therapies are promising clinical treatments for curing patients. However, efficiently identifying valid drug combinations remains challenging because the number of available drugs has increased rapidly. In this study, we proposed a deep learning model called the Dual Feature Fusion Network for Drug-Drug Synergy prediction (DFFNDDS) that utilizes a fine-tuned pretrained language model and dual feature fusion mechanism to predict synergistic drug combinations. The dual feature fusion mechanism fuses the drug features and cell line features at the bit-wise level and the vector-wise level. We demonstrated that DFFNDDS outperforms competitive methods and can serve as a reliable tool for identifying synergistic drug combinations.
Collapse
Affiliation(s)
- Mengdie Xu
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Xinwei Zhao
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Jingyu Wang
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Wei Feng
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Naifeng Wen
- School of Mechanical and Electrical Engineering, Dalian Minzu University, Dalian, China
| | - Chunyu Wang
- Faculty of Computing, Harbin Institute of Technology, Harbin, China
| | - Junjie Wang
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
- Institute of Medical Informatics and Management, Nanjing Medical University, No. 300 Guang Zhou Road, Nanjing, 210029 China
| | - Yun Liu
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
- Institute of Medical Informatics and Management, Nanjing Medical University, No. 300 Guang Zhou Road, Nanjing, 210029 China
- Department of Information, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Lingling Zhao
- Faculty of Computing, Harbin Institute of Technology, Harbin, China
| |
Collapse
|
35
|
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: 14] [Impact Index Per Article: 7.0] [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
|
36
|
Hosseini SR, Zhou X. CCSynergy: an integrative deep-learning framework enabling context-aware prediction of anti-cancer drug synergy. Brief Bioinform 2023; 24:bbac588. [PMID: 36562722 PMCID: PMC9851301 DOI: 10.1093/bib/bbac588] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/21/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
Combination therapy is a promising strategy for confronting the complexity of cancer. However, experimental exploration of the vast space of potential drug combinations is costly and unfeasible. Therefore, computational methods for predicting drug synergy are much needed for narrowing down this space, especially when examining new cellular contexts. Here, we thus introduce CCSynergy, a flexible, context aware and integrative deep-learning framework that we have established to unleash the potential of the Chemical Checker extended drug bioactivity profiles for the purpose of drug synergy prediction. We have shown that CCSynergy enables predictions of superior accuracy, remarkable robustness and improved context generalizability as compared to the state-of-the-art methods in the field. Having established the potential of CCSynergy for generating experimentally validated predictions, we next exhaustively explored the untested drug combination space. This resulted in a compendium of potentially synergistic drug combinations on hundreds of cancer cell lines, which can guide future experimental screens.
Collapse
Affiliation(s)
- Sayed-Rzgar Hosseini
- School of Biomedical Informatics, University of Texas Health Science Center (UTHealth), Houston, TX, USA
| | - Xiaobo Zhou
- School of Biomedical Informatics, University of Texas Health Science Center (UTHealth), Houston, TX, USA
| |
Collapse
|
37
|
She S, Chen H, Ji W, Sun M, Cheng J, Rui M, Feng C. Deep learning-based multi-drug synergy prediction model for individually tailored anti-cancer therapies. Front Pharmacol 2022; 13:1032875. [PMID: 36588694 PMCID: PMC9797718 DOI: 10.3389/fphar.2022.1032875] [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: 08/31/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
While synergistic drug combinations are more effective at fighting tumors with complex pathophysiology, preference compensating mechanisms, and drug resistance, the identification of novel synergistic drug combinations, especially complex higher-order combinations, remains challenging due to the size of combination space. Even though certain computational methods have been used to identify synergistic drug combinations in lieu of traditional in vitro and in vivo screening tests, the majority of previously published work has focused on predicting synergistic drug pairs for specific types of cancer and paid little attention to the sophisticated high-order combinations. The main objective of this study is to develop a deep learning-based approach that integrated multi-omics data to predict novel synergistic multi-drug combinations (DeepMDS) in a given cell line. To develop this approach, we firstly created a dataset comprising of gene expression profiles of cancer cell lines, target information of anti-cancer drugs, and drug response against a large variety of cancer cell lines. Based on the principle of a fully connected feed forward Deep Neural Network, the proposed model was constructed using this dataset, which achieved a high performance with a Mean Square Error (MSE) of 2.50 and a Root Mean Squared Error (RMSE) of 1.58 in the regression task, and gave the best classification accuracy of 0.94, an area under the Receiver Operating Characteristic curve (AUC) of 0.97, a sensitivity of 0.95, and a specificity of 0.93. Furthermore, we utilized three breast cancer cell subtypes (MCF-7, MDA-MD-468 and MDA-MB-231) and one lung cancer cell line A549 to validate the predicted results of our model, showing that the predicted top-ranked multi-drug combinations had superior anti-cancer effects to other combinations, particularly those that were widely used in clinical treatment. Our model has the potential to increase the practicality of expanding the drug combinational space and to leverage its capacity to prioritize the most effective multi-drug combinational therapy for precision oncology applications.
Collapse
Affiliation(s)
| | | | | | | | | | - Mengjie Rui
- *Correspondence: Chunlai Feng, ; Mengjie Rui,
| | | |
Collapse
|
38
|
Zhang P, Tu S, Zhang W, Xu L. Predicting cell line-specific synergistic drug combinations through a relational graph convolutional network with attention mechanism. Brief Bioinform 2022; 23:6711412. [PMID: 36136353 DOI: 10.1093/bib/bbac403] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 08/04/2022] [Accepted: 08/20/2022] [Indexed: 12/14/2022] Open
Abstract
Identifying synergistic drug combinations (SDCs) is a great challenge due to the combinatorial complexity and the fact that SDC is cell line specific. The existing computational methods either did not consider the cell line specificity of SDC, or did not perform well by building model for each cell line independently. In this paper, we present a novel encoder-decoder network named SDCNet for predicting cell line-specific SDCs. SDCNet learns common patterns across different cell lines as well as cell line-specific features in one model for drug combinations. This is realized by considering the SDC graphs of different cell lines as a relational graph, and constructing a relational graph convolutional network (R-GCN) as the encoder to learn and fuse the deep representations of drugs for different cell lines. An attention mechanism is devised to integrate the drug features from different layers of the R-GCN according to their relative importance so that representation learning is further enhanced. The common patterns are exploited through partial parameter sharing in cell line-specific decoders, which not only reconstruct the known SDCs but also predict new ones for each cell line. Experiments on various datasets demonstrate that SDCNet is superior to state-of-the-art methods and is also robust when generalized to new cell lines that are different from the training ones. Finally, the case study again confirms the effectiveness of our method in predicting novel reliable cell line-specific SDCs.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Computer Science and Engineering, Center for Cognitive Machines and Computational Health (CMaCH), Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shikui Tu
- Department of Computer Science and Engineering, Center for Cognitive Machines and Computational Health (CMaCH), Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wen Zhang
- Agricultural Bioinformatics Key Laboratory of Hubei Province, Hubei Engineering Technology Research Center of Agricultural Big Data, College of informatics, Huazhong Agricultural University, Wuhan 430070, China
| | - Lei Xu
- Department of Computer Science and Engineering, Center for Cognitive Machines and Computational Health (CMaCH), Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
39
|
Preto AJ, Matos-Filipe P, Mourão J, Moreira IS. SYNPRED: prediction of drug combination effects in cancer using different synergy metrics and ensemble learning. Gigascience 2022; 11:giac087. [PMID: 36155782 PMCID: PMC9511701 DOI: 10.1093/gigascience/giac087] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 06/14/2022] [Accepted: 08/18/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND In cancer research, high-throughput screening technologies produce large amounts of multiomics data from different populations and cell types. However, analysis of such data encounters difficulties due to disease heterogeneity, further exacerbated by human biological complexity and genomic variability. The specific profile of cancer as a disease (or, more realistically, a set of diseases) urges the development of approaches that maximize the effect while minimizing the dosage of drugs. Now is the time to redefine the approach to drug discovery, bringing an artificial intelligence (AI)-powered informational view that integrates the relevant scientific fields and explores new territories. RESULTS Here, we show SYNPRED, an interdisciplinary approach that leverages specifically designed ensembles of AI algorithms, as well as links omics and biophysical traits to predict anticancer drug synergy. It uses 5 reference models (Bliss, Highest Single Agent, Loewe, Zero Interaction Potency, and Combination Sensitivity Score), which, coupled with AI algorithms, allowed us to attain the ones with the best predictive performance and pinpoint the most appropriate reference model for synergy prediction, often overlooked in similar studies. By using an independent test set, SYNPRED exhibits state-of-the-art performance metrics either in the classification (accuracy, 0.85; precision, 0.91; recall, 0.90; area under the receiver operating characteristic, 0.80; and F1-score, 0.91) or in the regression models, mainly when using the Combination Sensitivity Score synergy reference model (root mean square error, 11.07; mean squared error, 122.61; Pearson, 0.86; mean absolute error, 7.43; Spearman, 0.87). Moreover, data interpretability was achieved by deploying the most current and robust feature importance approaches. A simple web-based application was constructed, allowing easy access by nonexpert researchers. CONCLUSIONS The performance of SYNPRED rivals that of the existing methods that tackle the same problem, yielding unbiased results trained with one of the most comprehensive datasets available (NCI ALMANAC). The leveraging of different reference models allowed deeper insights into which of them can be more appropriately used for synergy prediction. The Combination Sensitivity Score clearly stood out with improved performance among the full scope of surveyed approaches and synergy reference models. Furthermore, SYNPRED takes a particular focus on data interpretability, which has been in the spotlight lately when using the most advanced AI techniques.
Collapse
Affiliation(s)
- António J Preto
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão, 3030-789 Coimbra, Portugal
| | - Pedro Matos-Filipe
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Joana Mourão
- CNC—Center for Neuroscience and Cell Biology, CIBB—Center for Innovative Biomedicine and Biotechnology, 3004-504 Coimbra, Portugal
| | - Irina S Moreira
- Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CNC—Center for Neuroscience and Cell Biology, CIBB—Center for Innovative Biomedicine and Biotechnology, 3004-504 Coimbra, Portugal
| |
Collapse
|
40
|
Kuru HI, Tastan O, Cicek AE. MatchMaker: A Deep Learning Framework for Drug Synergy Prediction. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:2334-2344. [PMID: 34086576 DOI: 10.1109/tcbb.2021.3086702] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Drug combination therapies have been a viable strategy for the treatment of complex diseases such as cancer due to increased efficacy and reduced side effects. However, experimentally validating all possible combinations for synergistic interaction even with high-throughout screens is intractable due to vast combinatorial search space. Computational techniques can reduce the number of combinations to be evaluated experimentally by prioritizing promising candidates. We present MatchMaker that predicts drug synergy scores using drug chemical structure information and gene expression profiles of cell lines in a deep learning framework. For the first time, our model utilizes the largest known drug combination dataset to date, DrugComb. We compare the performance of MatchMaker with the state-of-the-art models and observe up to ∼ 15% correlation and ∼ 33% mean squared error (MSE) improvements over the next best method. We investigate the cell types and drug pairs that are relatively harder to predict and present novel candidate pairs. MatchMaker is built and available at https://github.com/tastanlab/matchmaker.
Collapse
|
41
|
Pinoli P, Ceddia G, Ceri S, Masseroli M. Predicting Drug Synergism by Means of Non-Negative Matrix Tri-Factorization. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:1956-1967. [PMID: 34166199 DOI: 10.1109/tcbb.2021.3091814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Traditional drug experiments to find synergistic drug pairs are time-consuming and expensive due to the numerous possible combinations of drugs that have to be examined. Thus, computational methods that can give suggestions for synergistic drug investigations are of great interest. Here, we propose a Non-negative Matrix Tri-Factorization (NMTF) based approach that leverages the integration of different data types for predicting synergistic drug pairs in multiple specific cell lines. Our computational framework relies on a network-based representation of available data about drug synergism, which also allows integrating genomic information about cell lines. We computationally evaluate the performances of our method in finding missing relationships between synergistic drug pairs and cell lines, and in computing synergy scores between drug pairs in a specific cell line, as well as we estimate the benefit of adding cell line genomic data to the network. Our approach obtains very good performance (Average Precision Score equal to 0.937, Pearson's correlation coefficient equal to 0.760) when cell line genomic data and rich data about synergistic drugs in a cell line are considered. Finally, we systematically searched our top-scored predictions in the available literature and in the NCI ALMANAC, a well-known database of drug combination experiments, proving the goodness of our findings.
Collapse
|
42
|
Sun M, She S, Chen H, Cheng J, Ji W, Wang D, Feng C. Prediction Model for Synergistic Anti-tumor Multi-compound Combinations from Traditional Chinese Medicine based on Extreme Gradient Boosting, Targets and Gene Expression Data. J Bioinform Comput Biol 2022; 20:2250016. [DOI: 10.1142/s0219720022500160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
43
|
Bhattacharjee S. Craft of Co-encapsulation in Nanomedicine: A Struggle To Achieve Synergy through Reciprocity. ACS Pharmacol Transl Sci 2022; 5:278-298. [PMID: 35592431 PMCID: PMC9112416 DOI: 10.1021/acsptsci.2c00033] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Indexed: 12/19/2022]
Abstract
Achieving synergism, often by combination therapy via codelivery of chemotherapeutic agents, remains the mainstay of treating multidrug-resistance cases in cancer and microbial strains. With a typical core-shell architecture and surface functionalization to ensure facilitated targeting of tissues, nanocarriers are emerging as a promising platform toward gaining such synergism. Co-encapsulation of disparate theranostic agents in nanocarriers-from chemotherapeutic molecules to imaging or photothermal modalities-can not only address the issue of protecting the labile drug payload from a hostile biochemical environment but may also ensure optimized drug release as a mainstay of synergistic effect. However, the fate of co-encapsulated molecules, influenced by temporospatial proximity, remains unpredictable and marred with events with deleterious impact on therapeutic efficacy, including molecular rearrangement, aggregation, and denaturation. Thus, more than just an art of confining multiple therapeutics into a 3D nanoscale space, a co-encapsulated nanocarrier, while aiming for synergism, should strive toward achieving a harmonious cohabitation of the encapsulated molecules that, despite proximity and opportunities for interaction, remain innocuous toward each other and ensure molecular integrity. This account will inspect the current progress in co-encapsulation in nanocarriers and distill out the key points toward accomplishing such synergism through reciprocity.
Collapse
Affiliation(s)
- Sourav Bhattacharjee
- School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
44
|
Rani P, Dutta K, Kumar V. Artificial intelligence techniques for prediction of drug synergy in malignant diseases: Past, present, and future. Comput Biol Med 2022; 144:105334. [DOI: 10.1016/j.compbiomed.2022.105334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 02/13/2022] [Accepted: 02/13/2022] [Indexed: 12/22/2022]
|
45
|
Torkamannia A, Omidi Y, Ferdousi R. A review of machine learning approaches for drug synergy prediction in cancer. Brief Bioinform 2022; 23:6552269. [PMID: 35323854 DOI: 10.1093/bib/bbac075] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/19/2022] [Accepted: 02/14/2022] [Indexed: 02/06/2023] Open
Abstract
Combinational pharmacotherapy with the synergistic/additive effect is a powerful treatment strategy for complex diseases such as malignancies. Identifying synergistic combinations with various compounds and structures requires testing a large number of compound combinations. However, in practice, examining different compounds by in vivo and in vitro approaches is costly, infeasible and challenging. In the last decades, significant success has been achieved by expanding computational methods in different pharmacological and bioinformatics domains. As promising tools, computational approaches such as machine learning algorithms (MLAs) are used for prioritizing combinational pharmacotherapies. This review aims to provide the models developed to predict synergistic drug combinations in cancer by MLAs with various information, including gene expression, protein-protein interactions, metabolite interactions, pathways and pharmaceutical information such as chemical structure, molecular descriptor and drug-target interactions.
Collapse
Affiliation(s)
- Anna Torkamannia
- Department of Health Information Technology, School of Management and Medical Informatics, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida, United States
| | - Reza Ferdousi
- Department of Health Information Technology, School of Management and Medical Informatics, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
46
|
Jaaks P, Coker EA, Vis DJ, Edwards O, Carpenter EF, Leto SM, Dwane L, Sassi F, Lightfoot H, Barthorpe S, van der Meer D, Yang W, Beck A, Mironenko T, Hall C, Hall J, Mali I, Richardson L, Tolley C, Morris J, Thomas F, Lleshi E, Aben N, Benes CH, Bertotti A, Trusolino L, Wessels L, Garnett MJ. Effective drug combinations in breast, colon and pancreatic cancer cells. Nature 2022; 603:166-173. [PMID: 35197630 PMCID: PMC8891012 DOI: 10.1038/s41586-022-04437-2] [Citation(s) in RCA: 200] [Impact Index Per Article: 66.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 01/18/2022] [Indexed: 02/08/2023]
Abstract
Combinations of anti-cancer drugs can overcome resistance and provide new treatments1,2. The number of possible drug combinations vastly exceeds what could be tested clinically. Efforts to systematically identify active combinations and the tissues and molecular contexts in which they are most effective could accelerate the development of combination treatments. Here we evaluate the potency and efficacy of 2,025 clinically relevant two-drug combinations, generating a dataset encompassing 125 molecularly characterized breast, colorectal and pancreatic cancer cell lines. We show that synergy between drugs is rare and highly context-dependent, and that combinations of targeted agents are most likely to be synergistic. We incorporate multi-omic molecular features to identify combination biomarkers and specify synergistic drug combinations and their active contexts, including in basal-like breast cancer, and microsatellite-stable or KRAS-mutant colon cancer. Our results show that irinotecan and CHEK1 inhibition have synergistic effects in microsatellite-stable or KRAS–TP53 double-mutant colon cancer cells, leading to apoptosis and suppression of tumour xenograft growth. This study identifies clinically relevant effective drug combinations in distinct molecular subpopulations and is a resource to guide rational efforts to develop combinatorial drug treatments. A survey of potency and efficacy of 2,025 clinically relevant two-drug combinations against 125 molecularly characterized breast, colorectal and pancreatic cancer cell lines identifies rare synergistic effects of anticancer drugs, informing rational combination treatments for specific cancer subtypes.
Collapse
Affiliation(s)
| | | | - Daniel J Vis
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Amsterdam, The Netherlands
| | | | | | | | - Lisa Dwane
- Wellcome Sanger Institute, Cambridge, UK
| | | | | | | | | | | | | | | | | | - James Hall
- Wellcome Sanger Institute, Cambridge, UK
| | - Iman Mali
- Wellcome Sanger Institute, Cambridge, UK
| | | | | | | | | | | | - Nanne Aben
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Cyril H Benes
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrea Bertotti
- Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy.,Department of Oncology, University of Torino School of Medicine, Turin, Italy
| | - Livio Trusolino
- Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy.,Department of Oncology, University of Torino School of Medicine, Turin, Italy
| | - Lodewyk Wessels
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of EEMCS, Delft University of Technology, Delft, The Netherlands.,Oncode Institute, Amsterdam, The Netherlands
| | | |
Collapse
|
47
|
Lin W, Wu L, Zhang Y, Wen Y, Yan B, Dai C, Liu K, He S, Bo X. An enhanced cascade-based deep forest model for drug combination prediction. Brief Bioinform 2022; 23:6513435. [DOI: 10.1093/bib/bbab562] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/20/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022] Open
Abstract
Abstract
Combination therapy has shown an obvious curative effect on complex diseases, whereas the search space of drug combinations is too large to be validated experimentally even with high-throughput screens. With the increase of the number of drugs, artificial intelligence techniques, especially machine learning methods, have become applicable for the discovery of synergistic drug combinations to significantly reduce the experimental workload. In this study, in order to predict novel synergistic drug combinations in various cancer cell lines, the cell line-specific drug-induced gene expression profile (GP) is added as a new feature type to capture the cellular response of drugs and reveal the biological mechanism of synergistic effect. Then, an enhanced cascade-based deep forest regressor (EC-DFR) is innovatively presented to apply the new small-scale drug combination dataset involving chemical, physical and biological (GP) properties of drugs and cells. Verified by the dataset, EC-DFR outperforms two state-of-the-art deep neural network-based methods and several advanced classical machine learning algorithms. Biological experimental validation performed subsequently on a set of previously untested drug combinations further confirms the performance of EC-DFR. What is more prominent is that EC-DFR can distinguish the most important features, making it more interpretable. By evaluating the contribution of each feature type, GP feature contributes 82.40%, showing the cellular responses of drugs may play crucial roles in synergism prediction. The analysis based on the top contributing genes in GP further demonstrates some potential relationships between the transcriptomic levels of key genes under drug regulation and the synergism of drug combinations.
Collapse
|
48
|
Xia F, Allen J, Balaprakash P, Brettin T, Garcia-Cardona C, Clyde A, Cohn J, Doroshow J, Duan X, Dubinkina V, Evrard Y, Fan YJ, Gans J, He S, Lu P, Maslov S, Partin A, Shukla M, Stahlberg E, Wozniak JM, Yoo H, Zaki G, Zhu Y, Stevens R. A cross-study analysis of drug response prediction in cancer cell lines. Brief Bioinform 2022; 23:bbab356. [PMID: 34524425 PMCID: PMC8769697 DOI: 10.1093/bib/bbab356] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/26/2021] [Accepted: 08/11/2021] [Indexed: 11/28/2022] Open
Abstract
To enable personalized cancer treatment, machine learning models have been developed to predict drug response as a function of tumor and drug features. However, most algorithm development efforts have relied on cross-validation within a single study to assess model accuracy. While an essential first step, cross-validation within a biological data set typically provides an overly optimistic estimate of the prediction performance on independent test sets. To provide a more rigorous assessment of model generalizability between different studies, we use machine learning to analyze five publicly available cell line-based data sets: National Cancer Institute 60, ancer Therapeutics Response Portal (CTRP), Genomics of Drug Sensitivity in Cancer, Cancer Cell Line Encyclopedia and Genentech Cell Line Screening Initiative (gCSI). Based on observed experimental variability across studies, we explore estimates of prediction upper bounds. We report performance results of a variety of machine learning models, with a multitasking deep neural network achieving the best cross-study generalizability. By multiple measures, models trained on CTRP yield the most accurate predictions on the remaining testing data, and gCSI is the most predictable among the cell line data sets included in this study. With these experiments and further simulations on partial data, two lessons emerge: (1) differences in viability assays can limit model generalizability across studies and (2) drug diversity, more than tumor diversity, is crucial for raising model generalizability in preclinical screening.
Collapse
Affiliation(s)
| | | | | | | | | | - Austin Clyde
- Argonne National Laboratory
- University of Chicago
| | | | | | | | | | | | - Ya Ju Fan
- Lawrence Livermore National Laboratory
| | | | | | - Pinyi Lu
- Frederick National Laboratory for Cancer Research
| | | | | | | | | | | | | | - George Zaki
- Frederick National Laboratory for Cancer Research
| | | | - Rick Stevens
- Argonne National Laboratory
- University of Chicago
| |
Collapse
|
49
|
Sukkar F, Shafaa M, El-Nagdy M, Darwish W. Polymeric Nanocarriers for Effective Synergistic Action of Sorafenib Tosylate and Gold-sensitized Gamma Radiation Against HepG2 Cells. Int J Nanomedicine 2022; 16:8309-8321. [PMID: 34992367 PMCID: PMC8711844 DOI: 10.2147/ijn.s331909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/21/2021] [Indexed: 12/17/2022] Open
Abstract
Purpose One of the key parameters towards effective and synergistic combinatorial anticancer therapeutic models is the nanocarrier. Nearly all previous studies have been limited to one nanocarrier for one drug. However, a comparative study on two polymeric nanocarriers for the same drug against the same cancer cell and under the same conditions helps to rationalize the properties of each polymeric nanocarrier to the effectiveness of the drug-loaded nanocapsules. Methods In this study, two of biocompatible polymers, namely poly lactic-co-glycolic acid (PLGA) and polyε-caprolactone (PCL), were used for co-delivery of sorafenib tosylate and gold nanoparticles (G). Results The anticancer effects of sorafenib tosylate (ST) combined with gold-sensitized radiation therapy were studied and rationalized to the physicochemical properties of each nanocarrier. Both models inhibited the proliferation of HepG2 cells via cell cycle arrest. The use of PCL and PLGA as nanocarriers for the proposed combined (chemo-radio) therapeutic model reduced the viability of HepG2 cells to 26% and 8%, respectively. PCL and PLGA models showed high necrosis levels (15.1 and 16.2, respectively). Conclusion Both PCL and PLGA are good nanocarriers for the proposed combined model. Compared to PCL NPs, PLGA NPs showed enhanced release, higher cytotoxicity and higher necrosis levels.
Collapse
Affiliation(s)
- Firas Sukkar
- Physics Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Medhat Shafaa
- Physics Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Mohamed El-Nagdy
- Physics Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Wael Darwish
- Department of Polymers and Pigments, National Research Centre, Giza, 12622, Egypt
| |
Collapse
|
50
|
Plana D, Palmer AC, Sorger PK. Independent Drug Action in Combination Therapy: Implications for Precision Oncology. Cancer Discov 2022; 12:606-624. [PMID: 34983746 DOI: 10.1158/2159-8290.cd-21-0212] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 09/02/2021] [Accepted: 11/10/2021] [Indexed: 11/16/2022]
Abstract
Combination therapies are superior to monotherapy for many cancers. This advantage was historically ascribed to the ability of combinations to address tumor heterogeneity, but synergistic interaction is now a common explanation as well as a design criterion for new combinations. We review evidence that independent drug action, described in 1961, explains the efficacy of many practice-changing combination therapies: it provides populations of patients with heterogeneous drug sensitivities multiple chances of benefit from at least one drug. Understanding response heterogeneity could reveal predictive or pharmacodynamic biomarkers for more precise use of existing drugs and realize the benefits of additivity or synergy.
Collapse
Affiliation(s)
- Deborah Plana
- Laboratory of Systems Pharmacology and the Department of Systems Biology, Harvard Medical School, Boston, Massachusetts
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts
| | - Adam C Palmer
- Department of Pharmacology, Computational Medicine Program, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Peter K Sorger
- Laboratory of Systems Pharmacology and the Department of Systems Biology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|