1
|
Huusari R, Wang T, Szedmak S, Dias D, Aittokallio T, Rousu J. Scaling up drug combination surface prediction. Brief Bioinform 2025; 26:bbaf099. [PMID: 40079263 PMCID: PMC11904408 DOI: 10.1093/bib/bbaf099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/15/2025] Open
Abstract
Drug combinations are required to treat advanced cancers and other complex diseases. Compared with monotherapy, combination treatments can enhance efficacy and reduce toxicity by lowering the doses of single drugs-and there especially synergistic combinations are of interest. Since drug combination screening experiments are costly and time-consuming, reliable machine learning models are needed for prioritizing potential combinations for further studies. Most of the current machine learning models are based on scalar-valued approaches, which predict individual response values or synergy scores for drug combinations. We take a functional output prediction approach, in which full, continuous dose-response combination surfaces are predicted for each drug combination on the cell lines. We investigate the predictive power of the recently proposed comboKR method, which is based on a powerful input-output kernel regression technique and functional modeling of the response surface. In this work, we develop a scaled-up formulation of the comboKR, which also implements improved modeling choices: we (1) incorporate new modeling choices for the output drug combination response surfaces to the comboKR framework, and (2) propose a projected gradient descent method to solve the challenging pre-image problem that is traditionally solved with simple candidate set approaches. We provide thorough experimental analysis of comboKR 2.0 with three real-word datasets within various challenging experimental settings, including cases where drugs or cell lines have not been encountered in the training data. Our comparison with synergy score prediction methods further highlights the relevance of dose-response prediction approaches, instead of relying on simple scoring methods.
Collapse
Affiliation(s)
- Riikka Huusari
- Department of Computer Science, Aalto University, Otakaari 1B, FI-00076 Espoo, Finland
| | - Tianduanyi Wang
- Department of Computer Science, Aalto University, Otakaari 1B, FI-00076 Espoo, Finland
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Tukholmankatu 8, FI-00270 Helsinki, Finland
| | - Sandor Szedmak
- Department of Computer Science, Aalto University, Otakaari 1B, FI-00076 Espoo, Finland
| | - Diogo Dias
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Tukholmankatu 8, FI-00270 Helsinki, Finland
- Hematology Research Unit, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 8, FI-00290 Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Haartmaninkatu 8, FI-00290 Helsinki, Finland
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Tukholmankatu 8, FI-00270 Helsinki, Finland
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Ullernchausseen 70, N-0379 Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Sognsvannsveien 9, N-0372 Oslo, Norway
| | - Juho Rousu
- Department of Computer Science, Aalto University, Otakaari 1B, FI-00076 Espoo, Finland
| |
Collapse
|
2
|
Chen J, Han H, Li L, Chen Z, Liu X, Li T, Wang X, Wang Q, Zhang R, Feng D, Yu L, Li X, Wang L, Li B, Li J. Prediction of cancer cell line-specific synergistic drug combinations based on multi-omics data. PeerJ 2025; 13:e19078. [PMID: 40028209 PMCID: PMC11869890 DOI: 10.7717/peerj.19078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 02/10/2025] [Indexed: 03/05/2025] Open
Abstract
Compared to single-drug therapy, combination therapy involves the use of two or more drugs to reduce drug dosage, decrease drug toxicity, and improve treatment efficacy. We developed an extreme gradient boosting (XGBoost)-based drug-drug cell line prediction model (XDDC) to predict synergistic drug combinations. XDDC was based on XGBoost and used one of the largest drug combination datasets, NCI-ALMANAC. In XDDC, drug chemical structures, adverse drug reactions, and target information were selected as drug features; gene expression, methylation, mutations, copy number variations, and RNA interference data were used as cell line features; and pathway information was incorporated to link drug features and cell line features. XDDC improved the interpretability of drug combination features and outperformed other machine learning methods. It achieved an area under the curve (AUC) of 0.966 ± 0.002 and an AUPR of 0.957 ± 0.002 when cross-validated on NCI-ALMANAC data. Different types of omics data were evaluated and compared in the model. Literature and experimental verification confirmed some of our predictions. XDDC could help medical professionals to rapidly screen synergistic drug combinations against specific cancer cell lines.
Collapse
Affiliation(s)
- Jiaqi Chen
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Huirui Han
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Lingxu Li
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Zhengxin Chen
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Xinying Liu
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Tianyi Li
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Xuefeng Wang
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Qibin Wang
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Ruijie Zhang
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Dehua Feng
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Lei Yu
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Xia Li
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Limei Wang
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Bing Li
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Jin Li
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
3
|
Ambreen S, Umar M, Noor A, Jain H, Ali R. Advanced AI and ML frameworks for transforming drug discovery and optimization: With innovative insights in polypharmacology, drug repurposing, combination therapy and nanomedicine. Eur J Med Chem 2025; 284:117164. [PMID: 39721292 DOI: 10.1016/j.ejmech.2024.117164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/24/2024] [Accepted: 11/27/2024] [Indexed: 12/28/2024]
Abstract
Artificial Intelligence (AI) and Machine Learning (ML) are transforming drug discovery by overcoming traditional challenges like high costs, time-consuming, and frequent failures. AI-driven approaches streamline key phases, including target identification, lead optimization, de novo drug design, and drug repurposing. Frameworks such as deep neural networks (DNNs), convolutional neural networks (CNNs), and deep reinforcement learning (DRL) models have shown promise in identifying drug targets, optimizing delivery systems, and accelerating drug repurposing. Generative adversarial networks (GANs) and variational autoencoders (VAEs) aid de novo drug design by creating novel drug-like compounds with desired properties. Case studies, such as DDR1 kinase inhibitors designed using generative models and CDK20 inhibitors developed via structure-based methods, highlight AI's ability to produce highly specific therapeutics. Models like SNF-CVAE and DeepDR further advance drug repurposing by uncovering new therapeutic applications for existing drugs. Advanced ML algorithms enhance precision in predicting drug efficacy, toxicity, and ADME-Tox properties, reducing development costs and improving drug-target interactions. AI also supports polypharmacology by optimizing multi-target drug interactions and enhances combination therapy through predictions of drug synergies and antagonisms. In nanomedicine, AI models like CURATE.AI and the Hartung algorithm optimize personalized treatments by predicting toxicological risks and real-time dosing adjustments with high accuracy. Despite its potential, challenges like data quality, model interpretability, and ethical concerns must be addressed. High-quality datasets, transparent models, and unbiased algorithms are essential for reliable AI applications. As AI continues to evolve, it is poised to revolutionize drug discovery and personalized medicine, advancing therapeutic development and patient care.
Collapse
Affiliation(s)
- Subiya Ambreen
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Mohammad Umar
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Aaisha Noor
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Himangini Jain
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Ruhi Ali
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India.
| |
Collapse
|
4
|
Tosh C, Tec M, White JB, Quinn JF, Ibanez Sanchez G, Calder P, Kung AL, Dela Cruz FS, Tansey W. A Bayesian active learning platform for scalable combination drug screens. Nat Commun 2025; 16:156. [PMID: 39746987 PMCID: PMC11696745 DOI: 10.1038/s41467-024-55287-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 12/05/2024] [Indexed: 01/04/2025] Open
Abstract
Large-scale combination drug screens are generally considered intractable due to the immense number of possible combinations. Existing approaches use ad hoc fixed experimental designs then train machine learning models to impute unobserved combinations. Here we propose BATCHIE, an orthogonal approach that conducts experiments dynamically in batches. BATCHIE uses information theory and probabilistic modeling to design each batch to be maximally informative based on the results of previous experiments. On retrospective experiments from previous large-scale screens, BATCHIE designs rapidly discover highly effective and synergistic combinations. In a prospective combination screen of a library of 206 drugs on a collection of pediatric cancer cell lines, the BATCHIE model accurately predicts unseen combinations and detects synergies after exploring only 4% of the 1.4M possible experiments. Further, the model identifies a panel of top combinations for Ewing sarcomas, which follow-up validation experiments confirm to be effective, including the rational and translatable top hit of PARP plus topoisomerase I inhibition. These results demonstrate that adaptive experiments can enable large-scale unbiased combination drug screens with a relatively small number of experiments. BATCHIE is open source and publicly available ( https://github.com/tansey-lab/batchie ).
Collapse
Affiliation(s)
- Christopher Tosh
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mauricio Tec
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jessica B White
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jeffrey F Quinn
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Paul Calder
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew L Kung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Filemon S Dela Cruz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wesley Tansey
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
5
|
Wang H, Li X, You X, Zhao G. Harnessing the power of artificial intelligence for human living organoid research. Bioact Mater 2024; 42:140-164. [PMID: 39280585 PMCID: PMC11402070 DOI: 10.1016/j.bioactmat.2024.08.027] [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: 04/30/2024] [Revised: 07/21/2024] [Accepted: 08/26/2024] [Indexed: 09/18/2024] Open
Abstract
As a powerful paradigm, artificial intelligence (AI) is rapidly impacting every aspect of our day-to-day life and scientific research through interdisciplinary transformations. Living human organoids (LOs) have a great potential for in vitro reshaping many aspects of in vivo true human organs, including organ development, disease occurrence, and drug responses. To date, AI has driven the revolutionary advances of human organoids in life science, precision medicine and pharmaceutical science in an unprecedented way. Herein, we provide a forward-looking review, the frontiers of LOs, covering the engineered construction strategies and multidisciplinary technologies for developing LOs, highlighting the cutting-edge achievements and the prospective applications of AI in LOs, particularly in biological study, disease occurrence, disease diagnosis and prediction and drug screening in preclinical assay. Moreover, we shed light on the new research trends harnessing the power of AI for LO research in the context of multidisciplinary technologies. The aim of this paper is to motivate researchers to explore organ function throughout the human life cycle, narrow the gap between in vitro microphysiological models and the real human body, accurately predict human-related responses to external stimuli (cues and drugs), accelerate the preclinical-to-clinical transformation, and ultimately enhance the health and well-being of patients.
Collapse
Affiliation(s)
- Hui Wang
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308, PR China
| | - Xiangyang Li
- Henan Engineering Research Center of Food Microbiology, College of food and bioengineering, Henan University of Science and Technology, Luoyang, 471023, PR China
- Haihe Laboratory of Synthetic Biology, Tianjin, 300308, PR China
| | - Xiaoyan You
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308, PR China
- Henan Engineering Research Center of Food Microbiology, College of food and bioengineering, Henan University of Science and Technology, Luoyang, 471023, PR China
| | - Guoping Zhao
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308, PR China
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032, PR China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- Engineering Laboratory for Nutrition, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, PR China
| |
Collapse
|
6
|
Jin I, Lee S, Schmuhalek M, Nam H. DD-PRiSM: a deep learning framework for decomposition and prediction of synergistic drug combinations. Brief Bioinform 2024; 26:bbae717. [PMID: 39800875 PMCID: PMC11725392 DOI: 10.1093/bib/bbae717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Combination therapies have emerged as a promising approach for treating complex diseases, particularly cancer. However, predicting the efficacy and safety profiles of these therapies remains a significant challenge, primarily because of the complex interactions among drugs and their wide-ranging effects. To address this issue, we introduce DD-PRiSM (Decomposition of Drug-Pair Response into Synergy and Monotherapy effect), a deep-learning pipeline that predicts the effects of combination therapy. DD-PRiSM consists of two predictive models. The first is the Monotherapy model, which predicts parameters of the drug response curve based on drug structure and cell line gene expression. This reconstructed curve is then used to predict cell viability at the given drug dosage. The second is the Combination therapy model, which predicts the efficacy of drug combinations by analyzing individual drug effects and their synergistic interactions with a specific dosage level of individual drugs. The efficacy of DD-PRiSM is demonstrated through its performance metrics, achieving a root mean square error of 0.0854, a Pearson correlation coefficient of 0.9063, and an R2 of 0.8209 for unseen pairs. Furthermore, DD-PRiSM distinguishes itself by its capability to decompose combination therapy efficacy, successfully identifying synergistic drug pairs. We demonstrated synergistic responses vary across cancer types and identified hub drugs that trigger synergistic effects. Finally, we suggested a promising drug pair through our case study.
Collapse
Affiliation(s)
- Iljung Jin
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology (GIST),Buk-gu, Gwangju 61005, Republic of Korea
| | - Songyeon Lee
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology (GIST),Buk-gu, Gwangju 61005, Republic of Korea
| | - Martin Schmuhalek
- AI Graduate School, Gwangju Institute of Science and Technology (GIST), Buk-gu, Gwangju 61005, Republic of Korea
| | - Hojung Nam
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology (GIST),Buk-gu, Gwangju 61005, Republic of Korea
- AI Graduate School, Gwangju Institute of Science and Technology (GIST), Buk-gu, Gwangju 61005, Republic of Korea
- Center for AI-Applied High Efficiency Drug Discovery (AHEDD), Gwangju Institute of Science and Technology (GIST), Buk-gu, Gwangju 61005, Republic of Korea
| |
Collapse
|
7
|
Velez-Arce A, Li MM, Gao W, Lin X, Huang K, Fu T, Pentelute BL, Kellis M, Zitnik M. Signals in the Cells: Multimodal and Contextualized Machine Learning Foundations for Therapeutics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598655. [PMID: 38948789 PMCID: PMC11212894 DOI: 10.1101/2024.06.12.598655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Drug discovery AI datasets and benchmarks have not traditionally included single-cell analysis biomarkers. While benchmarking efforts in single-cell analysis have recently released collections of single-cell tasks, they have yet to comprehensively release datasets, models, and benchmarks that integrate a broad range of therapeutic discovery tasks with cell-type-specific biomarkers. Therapeutics Commons (TDC-2) presents datasets, tools, models, and benchmarks integrating cell-type-specific contextual features with ML tasks across therapeutics. We present four tasks for contextual learning at single-cell resolution: drug-target nomination, genetic perturbation response prediction, chemical perturbation response prediction, and protein-peptide interaction prediction. We introduce datasets, models, and benchmarks for these four tasks. Finally, we detail the advancements and challenges in machine learning and biology that drove the implementation of TDC-2 and how they are reflected in its architecture, datasets and benchmarks, and foundation model tooling.
Collapse
Affiliation(s)
| | - Michelle M. Li
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115
| | - Wenhao Gao
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Xiang Lin
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115
| | - Kexin Huang
- Department of Computer Science, Stanford School of Engineering, Stanford, CA 94305
| | - Tianfan Fu
- Department of Computational Science, Rensselaer Polytechnic Institute, Troy, NY 12180
| | - Bradley L. Pentelute
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Manolis Kellis
- Broad Institute of MIT and Harvard, Computer Science and Artificial Intelligence Laboratory, MIT, Electrical Engineering and Computer Science Department, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Marinka Zitnik
- Broad Institute of MIT and Harvard, Harvard Data Science Initiative, Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02215
| |
Collapse
|
8
|
Wang Q, Liu X, Yan G. Predicting effective drug combinations for cancer treatment using a graph-based approach. Synth Syst Biotechnol 2024; 10:148-155. [PMID: 39469106 PMCID: PMC11513824 DOI: 10.1016/j.synbio.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/17/2024] [Accepted: 09/08/2024] [Indexed: 10/30/2024] Open
Abstract
Drug combination therapy, involving the use of two or more drugs, has been widely employed to treat complex diseases such as cancer. It enhances therapeutic efficacy, reduces drug resistance, and minimizes side effects. However, traditional methods to identify effective drug combinations are time-consuming, costly, and less efficient than computational methods. Therefore, developing computational approaches to predict drug combinations has become increasingly important. In this paper, we developed the Random Walk with Restart for Drug Combination (RWRDC) model to predict effective drug combinations for cancer therapy. The RWRDC model offers a quantitative mathematical method for predicting the potential effective drug combinations. Cross-validation results indicate that the RWRDC model outperforms other predictive models, particularly in breast, colorectal, and lung cancer predictions across various performance metrics. We have theoretically proven the convergence of its algorithm and provided an explanation for the algorithm's rationality. A targeted case study on breast cancer further highlights the capability of RWRDC to identify effective drug combinations. These findings highlight our model as a novel and effective tool for discovering potential effective drug combinations, offering new possibilities in therapy. Additionally, the graph-based framework of RWRDC holds potential for predicting drug combinations in other complex diseases, expanding its utility in the medical field.
Collapse
Affiliation(s)
- Qi Wang
- College of Science, China Agricultural University, Beijing, 100083, China
| | - Xiya Liu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Guiying Yan
- Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
9
|
Majidifar S, Zabihian A, Hooshmand M. Combination therapy synergism prediction for virus treatment using machine learning models. PLoS One 2024; 19:e0309733. [PMID: 39231124 PMCID: PMC11373828 DOI: 10.1371/journal.pone.0309733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 08/16/2024] [Indexed: 09/06/2024] Open
Abstract
Combining different drugs synergistically is an essential aspect of developing effective treatments. Although there is a plethora of research on computational prediction for new combination therapies, there is limited to no research on combination therapies in the treatment of viral diseases. This paper proposes AI-based models for predicting novel antiviral combinations to treat virus diseases synergistically. To do this, we assembled a comprehensive dataset comprising information on viral strains, drug compounds, and their known interactions. As far as we know, this is the first dataset and learning model on combination therapy for viruses. Our proposal includes using a random forest model, an SVM model, and a deep model to train viral combination therapy. The machine learning models showed the highest performance, and the predicted values were validated by a t-test, indicating the effectiveness of the proposed methods. One of the predicted combinations of acyclovir and ribavirin has been experimentally confirmed to have a synergistic antiviral effect against herpes simplex type-1 virus, as described in the literature.
Collapse
Affiliation(s)
- Shayan Majidifar
- Department of Computer Science and Information Technology, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran
| | - Arash Zabihian
- Department of QA, Kimia Zist Parsian Pharmaceutical Company, Zanjan, Iran
| | - Mohsen Hooshmand
- Department of Computer Science and Information Technology, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran
| |
Collapse
|
10
|
Jiang J, Wei X, Lu Y, Li S, Xu X. Network-based prediction of anti-cancer drug combinations. Front Pharmacol 2024; 15:1418902. [PMID: 39211773 PMCID: PMC11357946 DOI: 10.3389/fphar.2024.1418902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Drug combinations have emerged as a promising therapeutic approach in cancer treatment, aimed at overcoming drug resistance and improving the efficacy of monotherapy regimens. However, identifying effective drug combinations has traditionally been time-consuming and often dependent on chance discoveries. Therefore, there is an urgent need to explore alternative strategies to support experimental research. In this study, we propose network-based prediction models to identify potential drug combinations for 11 types of cancer. Our approach involves extracting 55,299 associations from literature and constructing human protein interactomes for each cancer type. To predict drug combinations, we measure the proximity of drug-drug relationships within the network and employ a correlation clustering framework to detect functional communities. Finally, we identify 61,754 drug combinations. Furthermore, we analyze the network configurations specific to different cancer types and identify 30 key genes and 21 pathways. The performance of these models is subsequently assessed through in vitro assays, which exhibit a significant level of agreement. These findings represent a valuable contribution to the development of network-based drug combination design strategies, presenting potential solutions to overcome drug resistance and enhance cancer treatment outcomes.
Collapse
Affiliation(s)
- Jue Jiang
- School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Xuxu Wei
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - YuKang Lu
- School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Simin Li
- School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Xue Xu
- School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
11
|
Abbasi F, Rousu J. New methods for drug synergy prediction: A mini-review. Curr Opin Struct Biol 2024; 86:102827. [PMID: 38705070 DOI: 10.1016/j.sbi.2024.102827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 04/12/2024] [Accepted: 04/12/2024] [Indexed: 05/07/2024]
Abstract
In this mini-review, we explore the new prediction methods for drug combination synergy relying on high-throughput combinatorial screens. The fast progress of the field is witnessed in the more than thirty original machine learning methods published since 2021, a clear majority of them based on deep learning techniques. We aim to put these articles under a unifying lens by highlighting the core technologies, the data sources, the input data types and synergy scores used in the methods, as well as the prediction scenarios and evaluation protocols that the articles deal with. Our finding is that the best methods accurately solve the synergy prediction scenarios involving known drugs or cell lines while the scenarios involving new drugs or cell lines still fall short of an accurate prediction level.
Collapse
Affiliation(s)
- Fatemeh Abbasi
- Laboratory of Bioinformatics and Drug Design (LBD), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Juho Rousu
- Department of Computer Science, Aalto University, Espoo, Finland.
| |
Collapse
|
12
|
Huangfu X, Zhang C, Li H, Li S, Li Y. SNSynergy: Similarity network-based machine learning framework for synergy prediction towards new cell lines and new anticancer drug combinations. Comput Biol Chem 2024; 110:108054. [PMID: 38522389 DOI: 10.1016/j.compbiolchem.2024.108054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 02/22/2024] [Accepted: 03/16/2024] [Indexed: 03/26/2024]
Abstract
The computational method has been proven to be a promising means for pre-screening large-scale anticancer drug combinations to support precision oncology applications. Pioneering efforts have been made to develop machine learning technology for predicting drug synergy, but high computational cost for training models as well as great diversity and limited size in screening data escalate the difficulty of prediction. To address this challenge, we propose a simple machine learning framework, namely Similarity Network-based Synergy prediction (SNSynergy), for predicting synergistic effects towards new cell lines and new drug combinations by two locally weighted models CLSN and DCSN. This framework only requires a small amount of auxiliary data, like genomics information of cell lines and the molecular fingerprints or targets of drugs. Based on the assumption that similar cell lines and similar drug combinations have similar synergistic effects, CLSN and DCSN predict synergy scores through capturing individual synergy contributions of nearest cell line and drug combination neighbors, respectively. High correlations between predicted and measured synergy scores on two leading cancer cell line pharmacogenomic screening datasets (the O'Neil dataset and the NCI-ALMANAC dataset) demonstrate the effectiveness and robustness of SNSynergy. Many of the identified drug combinations are consistent with previous studies, or have been explored in clinical settings against the specific cancer type, showing that SNSynergy has the potential to supply cost-saving and effective high-throughput screening for prioritizing the most applicable cell lines and the most promising drug combinations.
Collapse
Affiliation(s)
- Xiaosheng Huangfu
- School of Science, Yanshan University, Qinhuangdao, Hebei 066004, China
| | - Chengwei Zhang
- School of Science, Yanshan University, Qinhuangdao, Hebei 066004, China
| | - Hualong Li
- School of Science, Yanshan University, Qinhuangdao, Hebei 066004, China; School of mathematics and statistics, Guangdong University of Technology, Guangzhou 510520, China
| | - Sile Li
- School of Science, Yanshan University, Qinhuangdao, Hebei 066004, China
| | - Yushuang Li
- School of Science, Yanshan University, Qinhuangdao, Hebei 066004, China.
| |
Collapse
|
13
|
Zhao X, Xu J, Shui Y, Xu M, Hu J, Liu X, Che K, Wang J, Liu Y. PermuteDDS: a permutable feature fusion network for drug-drug synergy prediction. J Cheminform 2024; 16:41. [PMID: 38622663 PMCID: PMC11017561 DOI: 10.1186/s13321-024-00839-8] [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: 09/19/2023] [Accepted: 04/03/2024] [Indexed: 04/17/2024] Open
Abstract
MOTIVATION Drug combination therapies have shown promise in clinical cancer treatments. However, it is hard to experimentally identify all drug combinations for synergistic interaction even with high-throughput screening due to the vast space of potential combinations. Although a number of computational methods for drug synergy prediction have proven successful in narrowing down this space, fusing drug pairs and cell line features effectively still lacks study, hindering current algorithms from understanding the complex interaction between drugs and cell lines. RESULTS In this paper, we proposed a Permutable feature fusion network for Drug-Drug Synergy prediction, named PermuteDDS. PermuteDDS takes multiple representations of drugs and cell lines as input and employs a permutable fusion mechanism to combine drug and cell line features. In experiments, PermuteDDS exhibits state-of-the-art performance on two benchmark data sets. Additionally, the results on independent test set grouped by different tissues reveal that PermuteDDS has good generalization performance. We believed that PermuteDDS is an effective and valuable tool for identifying synergistic drug combinations. It is publicly available at https://github.com/littlewei-lazy/PermuteDDS . SCIENTIFIC CONTRIBUTION First, this paper proposes a permutable feature fusion network for predicting drug synergy termed PermuteDDS, which extract diverse information from multiple drug representations and cell line representations. Second, the permutable fusion mechanism combine the drug and cell line features by integrating information of different channels, enabling the utilization of complex relationships between drugs and cell lines. Third, comparative and ablation experiments provide evidence of the efficacy of PermuteDDS in predicting drug-drug synergy.
Collapse
Affiliation(s)
- Xinwei Zhao
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, China
| | - Junqing Xu
- The Second Clinical Medical School, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, China
| | - Youyuan Shui
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, China
| | - Mengdie Xu
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, China
| | - Jie Hu
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, China
- Institute of Medical Informatics and Management, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 210029, Jiangsu, China
| | - Xiaoyan Liu
- Faculty of Computing, Harbin Institute of Technology, No. 92 West Da Zhi St, Harbin, 150001, Heilongjiang, China
| | - Kai Che
- Xi'an Aeronautics Computing Technique Research Institute, AVIC, No. 156, TaiBai Nroth Road, Xi'an, 710068, Shanxi, China
- Aviation Key Laboratory of Science and Technology on Airborne and Missleborne Computer, Xi'an, 710065, Shanxi, China
| | - Junjie Wang
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, China.
- Institute of Medical Informatics and Management, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 210029, Jiangsu, China.
| | - Yun Liu
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, China.
- Institute of Medical Informatics and Management, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 210029, Jiangsu, China.
- Department of Information, the First Affiliated Hospital, Nanjing Medical University, No. 300 Guang Zhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
14
|
Dong Y, Chang Y, Wang Y, Han Q, Wen X, Yang Z, Zhang Y, Qiang Y, Wu K, Fan X, Ren X. MFSynDCP: multi-source feature collaborative interactive learning for drug combination synergy prediction. BMC Bioinformatics 2024; 25:140. [PMID: 38561679 PMCID: PMC10985899 DOI: 10.1186/s12859-024-05765-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 03/28/2024] [Indexed: 04/04/2024] Open
Abstract
Drug combination therapy is generally more effective than monotherapy in the field of cancer treatment. However, screening for effective synergistic combinations from a wide range of drug combinations is particularly important given the increase in the number of available drug classes and potential drug-drug interactions. Existing methods for predicting the synergistic effects of drug combinations primarily focus on extracting structural features of drug molecules and cell lines, but neglect the interaction mechanisms between cell lines and drug combinations. Consequently, there is a deficiency in comprehensive understanding of the synergistic effects of drug combinations. To address this issue, we propose a drug combination synergy prediction model based on multi-source feature interaction learning, named MFSynDCP, aiming to predict the synergistic effects of anti-tumor drug combinations. This model includes a graph aggregation module with an adaptive attention mechanism for learning drug interactions and a multi-source feature interaction learning controller for managing information transfer between different data sources, accommodating both drug and cell line features. Comparative studies with benchmark datasets demonstrate MFSynDCP's superiority over existing methods. Additionally, its adaptive attention mechanism graph aggregation module identifies drug chemical substructures crucial to the synergy mechanism. Overall, MFSynDCP is a robust tool for predicting synergistic drug combinations. The source code is available from GitHub at https://github.com/kkioplkg/MFSynDCP .
Collapse
Affiliation(s)
- Yunyun Dong
- School of Software, Taiyuan University of Technology, Taiyuan, Shanxi, China.
| | - Yunqing Chang
- School of Software, Taiyuan University of Technology, Taiyuan, Shanxi, China
| | - Yuxiang Wang
- School of Software, Taiyuan University of Technology, Taiyuan, Shanxi, China
| | - Qixuan Han
- School of Software, Taiyuan University of Technology, Taiyuan, Shanxi, China
| | - Xiaoyuan Wen
- School of Software, Taiyuan University of Technology, Taiyuan, Shanxi, China
| | - Ziting Yang
- School of Software, Taiyuan University of Technology, Taiyuan, Shanxi, China
| | - Yan Zhang
- School of Software, Taiyuan University of Technology, Taiyuan, Shanxi, China
| | - Yan Qiang
- College of Computer Science and Technology (College of Data Science), Taiyuan University of Technology, Taiyuan, Shanxi, China.
| | - Kun Wu
- School of Computing, University of Leeds, Leeds, West Yorkshire, UK
| | - Xiaole Fan
- Information Management Department, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Xiaoqiang Ren
- Information Management Department, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| |
Collapse
|
15
|
Nguyen T, Campbell A, Kumar A, Amponsah E, Fiterau M, Shahriyari L. Optimal fusion of genotype and drug embeddings in predicting cancer drug response. Brief Bioinform 2024; 25:bbae227. [PMID: 38754407 PMCID: PMC11097979 DOI: 10.1093/bib/bbae227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 04/14/2024] [Accepted: 04/25/2024] [Indexed: 05/18/2024] Open
Abstract
Predicting cancer drug response using both genomics and drug features has shown some success compared to using genomics features alone. However, there has been limited research done on how best to combine or fuse the two types of features. Using a visible neural network with two deep learning branches for genes and drug features as the base architecture, we experimented with different fusion functions and fusion points. Our experiments show that injecting multiplicative relationships between gene and drug latent features into the original concatenation-based architecture DrugCell significantly improved the overall predictive performance and outperformed other baseline models. We also show that different fusion methods respond differently to different fusion points, indicating that the relationship between drug features and different hierarchical biological level of gene features is optimally captured using different methods. Considering both predictive performance and runtime speed, tensor product partial is the best-performing fusion function to combine late-stage representations of drug and gene features to predict cancer drug response.
Collapse
Affiliation(s)
- Trang Nguyen
- Department of Computer Science, University of Massachusetts Amherst, Amherst 01002, MA, United States
| | - Anthony Campbell
- Department of Computer Science, University of Massachusetts Amherst, Amherst 01002, MA, United States
| | - Ankit Kumar
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst 01002, MA, United States
| | - Edwin Amponsah
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst 01002, MA, United States
| | - Madalina Fiterau
- Department of Computer Science, University of Massachusetts Amherst, Amherst 01002, MA, United States
| | - Leili Shahriyari
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst 01002, MA, United States
| |
Collapse
|
16
|
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
|
17
|
Joisa CU, Chen KA, Beville S, Stuhlmiller T, Berginski ME, Okumu D, Golitz BT, East MP, Johnson GL, Gomez SM. Combined kinome inhibition states are predictive of cancer cell line sensitivity to kinase inhibitor combination therapies. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2024; 29:276-290. [PMID: 38160286 PMCID: PMC11413988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Protein kinases are a primary focus in targeted therapy development for cancer, owing to their role as regulators in nearly all areas of cell life. Recent strategies targeting the kinome with combination therapies have shown promise, such as trametinib and dabrafenib in advanced melanoma, but empirical design for less characterized pathways remains a challenge. Computational combination screening is an attractive alternative, allowing in-silico filtering prior to experimental testing of drastically fewer leads, increasing efficiency and effectiveness of drug development pipelines. In this work, we generated combined kinome inhibition states of 40,000 kinase inhibitor combinations from kinobeads-based kinome profiling across 64 doses. We then integrated these with transcriptomics from CCLE to build machine learning models with elastic-net feature selection to predict cell line sensitivity across nine cancer types, with accuracy R2 ∼ 0.75-0.9. We then validated the model by using a PDX-derived TNBC cell line and saw good global accuracy (R2 ∼ 0.7) as well as high accuracy in predicting synergy using four popular metrics (R2 ∼ 0.9). Additionally, the model was able to predict a highly synergistic combination of trametinib and omipalisib for TNBC treatment, which incidentally was recently in phase I clinical trials. Our choice of tree-based models for greater interpretability allowed interrogation of highly predictive kinases in each cancer type, such as the MAPK, CDK, and STK kinases. Overall, these results suggest that kinome inhibition states of kinase inhibitor combinations are strongly predictive of cell line responses and have great potential for integration into computational drug screening pipelines. This approach may facilitate the identification of effective kinase inhibitor combinations and accelerate the development of novel cancer therapies, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Chinmaya U Joisa
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA2North Carolina State University, Raleigh, NC, USA3Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Pellegrini M. Advances in Network-Based Drug Repositioning. LECTURE NOTES IN COMPUTER SCIENCE 2024:99-114. [DOI: 10.1007/978-3-031-55248-9_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
19
|
Yuan J, Zhang Y, Wang X. Application of machine learning in the management of lymphoma: Current practice and future prospects. Digit Health 2024; 10:20552076241247963. [PMID: 38628632 PMCID: PMC11020711 DOI: 10.1177/20552076241247963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/28/2024] [Indexed: 04/19/2024] Open
Abstract
In the past decade, digitization of medical records and multiomics data analysis in lymphoma has led to the accessibility of high-dimensional records. The digitization of medical records, the visualization of extensive volume data extracted from medical images, and the integration of multiomics methods into clinical decision-making have produced many datasets. As a promising auxiliary tool, machine learning (ML) intends to extract homologous features in large-scale data sets and encode them into various patterns to complete complicated tasks. At present, artificial intelligence and digital mining have shown promising prospects in the field of lymphoma pathological image analysis. The paradigm shift from qualitative analysis to quantitative analysis makes the pathological diagnosis more intelligent and the results more accurate and objective. ML can promote accurate lymphoma diagnosis and provide patients with prognostic information and more individualized treatment options. Based on the above, this comprehensive review of the general workflow of ML highlights recent advances in ML techniques in the diagnosis, treatment, and prognosis of lymphoma, and clarifies the boundedness and future orientation of the ML technique in the clinical practice of lymphoma.
Collapse
Affiliation(s)
- Junyun Yuan
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Ya Zhang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Taishan Scholars Program of Shandong Province, Jinan, Shandong, China
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Taishan Scholars Program of Shandong Province, Jinan, Shandong, China
- Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong, China
- National Clinical Research Center for Hematologic Diseases, Hospital of Soochow University, Suzhou, China
| |
Collapse
|
20
|
Chen D, Wang X, Zhu H, Jiang Y, Li Y, Liu Q, Liu Q. Predicting anticancer synergistic drug combinations based on multi-task learning. BMC Bioinformatics 2023; 24:448. [PMID: 38012551 PMCID: PMC10680313 DOI: 10.1186/s12859-023-05524-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 10/09/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND The discovery of anticancer drug combinations is a crucial work of anticancer treatment. In recent years, pre-screening drug combinations with synergistic effects in a large-scale search space adopting computational methods, especially deep learning methods, is increasingly popular with researchers. Although achievements have been made to predict anticancer synergistic drug combinations based on deep learning, the application of multi-task learning in this field is relatively rare. The successful practice of multi-task learning in various fields shows that it can effectively learn multiple tasks jointly and improve the performance of all the tasks. METHODS In this paper, we propose MTLSynergy which is based on multi-task learning and deep neural networks to predict synergistic anticancer drug combinations. It simultaneously learns two crucial prediction tasks in anticancer treatment, which are synergy prediction of drug combinations and sensitivity prediction of monotherapy. And MTLSynergy integrates the classification and regression of prediction tasks into the same model. Moreover, autoencoders are employed to reduce the dimensions of input features. RESULTS Compared with the previous methods listed in this paper, MTLSynergy achieves the lowest mean square error of 216.47 and the highest Pearson correlation coefficient of 0.76 on the drug synergy prediction task. On the corresponding classification task, the area under the receiver operator characteristics curve and the area under the precision-recall curve are 0.90 and 0.62, respectively, which are equivalent to the comparison methods. Through the ablation study, we verify that multi-task learning and autoencoder both have a positive effect on prediction performance. In addition, the prediction results of MTLSynergy in many cases are also consistent with previous studies. CONCLUSION Our study suggests that multi-task learning is significantly beneficial for both drug synergy prediction and monotherapy sensitivity prediction when combining these two tasks into one model. The ability of MTLSynergy to discover new anticancer synergistic drug combinations noteworthily outperforms other state-of-the-art methods. MTLSynergy promises to be a powerful tool to pre-screen anticancer synergistic drug combinations.
Collapse
Affiliation(s)
- Danyi Chen
- School of Software Engineering, Tongji University, Shanghai, 201804, China
| | - Xiaowen Wang
- School of Software Engineering, Tongji University, Shanghai, 201804, China
| | - Hongming Zhu
- School of Software Engineering, Tongji University, Shanghai, 201804, China
| | - Yizhi Jiang
- School of Software Engineering, Tongji University, Shanghai, 201804, China
| | - Yulong Li
- School of Software Engineering, Tongji University, Shanghai, 201804, China
| | - Qi Liu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Qin Liu
- School of Software Engineering, Tongji University, Shanghai, 201804, China.
| |
Collapse
|
21
|
Singh N, Khan FM, Bala L, Vera J, Wolkenhauer O, Pützer B, Logotheti S, Gupta SK. Logic-based modeling and drug repurposing for the prediction of novel therapeutic targets and combination regimens against E2F1-driven melanoma progression. BMC Chem 2023; 17:161. [PMID: 37993971 PMCID: PMC10666365 DOI: 10.1186/s13065-023-01082-2] [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: 06/12/2023] [Accepted: 11/08/2023] [Indexed: 11/24/2023] Open
Abstract
Melanoma presents increasing prevalence and poor outcomes. Progression to aggressive stages is characterized by overexpression of the transcription factor E2F1 and activation of downstream prometastatic gene regulatory networks (GRNs). Appropriate therapeutic manipulation of the E2F1-governed GRNs holds the potential to prevent metastasis however, these networks entail complex feedback and feedforward regulatory motifs among various regulatory layers, which make it difficult to identify druggable components. To this end, computational approaches such as mathematical modeling and virtual screening are important tools to unveil the dynamics of these signaling networks and identify critical components that could be further explored as therapeutic targets. Herein, we integrated a well-established E2F1-mediated epithelial-mesenchymal transition (EMT) map with transcriptomics data from E2F1-expressing melanoma cells to reconstruct a core regulatory network underlying aggressive melanoma. Using logic-based in silico perturbation experiments of a core regulatory network, we identified that simultaneous perturbation of Protein kinase B (AKT1) and oncoprotein murine double minute 2 (MDM2) drastically reduces EMT in melanoma. Using the structures of the two protein signatures, virtual screening strategies were performed with the FDA-approved drug library. Furthermore, by combining drug repurposing and computer-aided drug design techniques, followed by molecular dynamics simulation analysis, we identified two potent drugs (Tadalafil and Finasteride) that can efficiently inhibit AKT1 and MDM2 proteins. We propose that these two drugs could be considered for the development of therapeutic strategies for the management of aggressive melanoma.
Collapse
Affiliation(s)
- Nivedita Singh
- Department of Biochemistry, BBDCODS, BBD University, Lucknow, Uttar Pradesh, India
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Faiz M Khan
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, Germany
| | - Lakshmi Bala
- Department of Biochemistry, BBDCODS, BBD University, Lucknow, Uttar Pradesh, India
| | - Julio Vera
- Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, Germany
- Leibniz Institute for Food Systems Biology, Technical University of Munich, Munich, Germany
- Chhattisgarh Swami Vivekanand Technical University, Bhilai, Chhattisgarh, India
- Stellenbosch Institute of Advanced Study, Wallenberg Research Centre, Stellenbosch University, Stellenbosch, South Africa
| | - Brigitte Pützer
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, Rostock, Germany
| | - Stella Logotheti
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, Rostock, Germany
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou, Athens, Greece
| | - Shailendra K Gupta
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, Germany.
- Chhattisgarh Swami Vivekanand Technical University, Bhilai, Chhattisgarh, India.
| |
Collapse
|
22
|
Wang W, Yuan G, Wan S, Zheng Z, Liu D, Zhang H, Li J, Zhou Y, Wang X. A granularity-level information fusion strategy on hypergraph transformer for predicting synergistic effects of anticancer drugs. Brief Bioinform 2023; 25:bbad522. [PMID: 38243692 PMCID: PMC10796255 DOI: 10.1093/bib/bbad522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 11/08/2023] [Accepted: 12/19/2023] [Indexed: 01/21/2024] Open
Abstract
Combination therapy has exhibited substantial potential compared to monotherapy. However, due to the explosive growth in the number of cancer drugs, the screening of synergistic drug combinations has become both expensive and time-consuming. Synergistic drug combinations refer to the concurrent use of two or more drugs to enhance treatment efficacy. Currently, numerous computational methods have been developed to predict the synergistic effects of anticancer drugs. However, there has been insufficient exploration of how to mine drug and cell line data at different granularity levels for predicting synergistic anticancer drug combinations. Therefore, this study proposes a granularity-level information fusion strategy based on the hypergraph transformer, named HypertranSynergy, to predict synergistic effects of anticancer drugs. HypertranSynergy introduces synergistic connections between cancer cell lines and drug combinations using hypergraph. Then, the Coarse-grained Information Extraction (CIE) module merges the hypergraph with a transformer for node embeddings. In the CIE module, Contranorm is a normalization layer that mitigates over-smoothing, while Gaussian noise addresses local information gaps. Additionally, the Fine-grained Information Extraction (FIE) module assesses fine-grained information's impact on predictions by employing similarity-aware matrices from drug/cell line features. Both CIE and FIE modules are integrated into HypertranSynergy. In addition, HypertranSynergy achieved the AUC of 0.93${\pm }$0.01 and the AUPR of 0.69${\pm }$0.02 in 5-fold cross-validation of classification task, and the RMSE of 13.77${\pm }$0.07 and the PCC of 0.81${\pm }$0.02 in 5-fold cross-validation of regression task. These results are better than most of the state-of-the-art models.
Collapse
Affiliation(s)
- Wei Wang
- College of Computer and Information Engineering, Henan Normal University, 453007 Xinxiang, China
- Key Laboratory of Artificial Intelligence and Personalized Learning in Education of Henan Province 453007, China
| | - Gaolin Yuan
- College of Computer and Information Engineering, Henan Normal University, 453007 Xinxiang, China
| | - Shitong Wan
- College of Computer and Information Engineering, Henan Normal University, 453007 Xinxiang, China
| | - Ziwei Zheng
- College of Computer and Information Engineering, Henan Normal University, 453007 Xinxiang, China
| | - Dong Liu
- College of Computer and Information Engineering, Henan Normal University, 453007 Xinxiang, China
- Key Laboratory of Artificial Intelligence and Personalized Learning in Education of Henan Province 453007, China
| | - Hongjun Zhang
- Hebi Instiute of Engineering and Technology, Henan Polytechnic University, 458030, China
| | - Juntao Li
- School of Mathematics and Information Science, Henan Normal University, 453007 Xinxiang, China
| | - Yun Zhou
- College of Computer and Information Engineering, Henan Normal University, 453007 Xinxiang, China
- Key Laboratory of Artificial Intelligence and Personalized Learning in Education of Henan Province 453007, China
| | - Xianfang Wang
- College of Computer Science and Technology Engineering, Henan Institute of Technology, 453000, China
| |
Collapse
|
23
|
Bertin P, Rector-Brooks J, Sharma D, Gaudelet T, Anighoro A, Gross T, Martínez-Peña F, Tang EL, Suraj MS, Regep C, Hayter JBR, Korablyov M, Valiante N, van der Sloot A, Tyers M, Roberts CES, Bronstein MM, Lairson LL, Taylor-King JP, Bengio Y. RECOVER identifies synergistic drug combinations in vitro through sequential model optimization. CELL REPORTS METHODS 2023; 3:100599. [PMID: 37797618 PMCID: PMC10626197 DOI: 10.1016/j.crmeth.2023.100599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 08/30/2023] [Accepted: 09/06/2023] [Indexed: 10/07/2023]
Abstract
For large libraries of small molecules, exhaustive combinatorial chemical screens become infeasible to perform when considering a range of disease models, assay conditions, and dose ranges. Deep learning models have achieved state-of-the-art results in silico for the prediction of synergy scores. However, databases of drug combinations are biased toward synergistic agents and results do not generalize out of distribution. During 5 rounds of experimentation, we employ sequential model optimization with a deep learning model to select drug combinations increasingly enriched for synergism and active against a cancer cell line-evaluating only ∼5% of the total search space. Moreover, we find that learned drug embeddings (using structural information) begin to reflect biological mechanisms. In silico benchmarking suggests search queries are ∼5-10× enriched for highly synergistic drug combinations by using sequential rounds of evaluation when compared with random selection or ∼3× when using a pretrained model.
Collapse
Affiliation(s)
- Paul Bertin
- Mila, the Quebec AI Institute, Montreal, QC, Canada
| | | | | | | | | | | | | | - Eileen L Tang
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | | | | | | | | | | | - Almer van der Sloot
- IRIC, Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, QC, Canada
| | - Mike Tyers
- Program in Molecular Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | | | - Michael M Bronstein
- Relation Therapeutics, London, UK; Department of Computer Science, University of Oxford, Oxford, UK
| | - Luke L Lairson
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | | | | |
Collapse
|
24
|
Glover HJ, Holliday H, Shparberg RA, Winkler D, Day M, Morris MB. Signalling pathway crosstalk stimulated by L-proline drives mouse embryonic stem cells to primitive-ectoderm-like cells. Development 2023; 150:dev201704. [PMID: 37823343 PMCID: PMC10652046 DOI: 10.1242/dev.201704] [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: 02/14/2023] [Accepted: 10/02/2023] [Indexed: 10/13/2023]
Abstract
The amino acid L-proline exhibits growth factor-like properties during development - from improving blastocyst development to driving neurogenesis in vitro. Addition of 400 μM L-proline to self-renewal medium drives naïve mouse embryonic stem cells (ESCs) to early primitive ectoderm-like (EPL) cells - a transcriptionally distinct primed or partially primed pluripotent state. EPL cells retain expression of pluripotency genes, upregulate primitive ectoderm markers, undergo a morphological change and have increased cell number. These changes are facilitated by a complex signalling network hinging on the Mapk, Fgfr, Pi3k and mTor pathways. Here, we use a factorial experimental design coupled with statistical modelling to understand which signalling pathways are involved in the transition between ESCs and EPL cells, and how they underpin changes in morphology, cell number, apoptosis, proliferation and gene expression. This approach reveals pathways which work antagonistically or synergistically. Most properties were affected by more than one inhibitor, and each inhibitor blocked specific aspects of the naïve-to-primed transition. These mechanisms underpin progression of stem cells across the in vitro pluripotency continuum and serve as a model for pre-, peri- and post-implantation embryogenesis.
Collapse
Affiliation(s)
- Hannah J. Glover
- School of Medical Sciences, University of Sydney, Sydney 2006, Australia
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Holly Holliday
- School of Medical Sciences, University of Sydney, Sydney 2006, Australia
| | | | - David Winkler
- Department of Biochemistry and Chemistry, Latrobe Institute for Molecular Science, Latrobe University, Bundoora 3083, Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Australia
- Advanced Materials and Healthcare Technologies, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Margot Day
- School of Medical Sciences, University of Sydney, Sydney 2006, Australia
| | - Michael B. Morris
- School of Medical Sciences, University of Sydney, Sydney 2006, Australia
| |
Collapse
|
25
|
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
|
26
|
Flanary VL, Fisher JL, Wilk EJ, Howton TC, Lasseigne BN. Computational Advancements in Cancer Combination Therapy Prediction. JCO Precis Oncol 2023; 7:e2300261. [PMID: 37824797 PMCID: PMC12012855 DOI: 10.1200/po.23.00261] [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: 05/24/2023] [Revised: 07/20/2023] [Accepted: 08/15/2023] [Indexed: 10/14/2023] Open
Abstract
Given the high attrition rate of de novo drug discovery and limited efficacy of single-agent therapies in cancer treatment, combination therapy prediction through in silico drug repurposing has risen as a time- and cost-effective alternative for identifying novel and potentially efficacious therapies for cancer. The purpose of this review is to provide an introduction to computational methods for cancer combination therapy prediction and to summarize recent studies that implement each of these methods. A systematic search of the PubMed database was performed, focusing on studies published within the past 10 years. Our search included reviews and articles of ongoing and retrospective studies. We prioritized articles with findings that suggest considerations for improving combination therapy prediction methods over providing a meta-analysis of all currently available cancer combination therapy prediction methods. Computational methods used for drug combination therapy prediction in cancer research include networks, regression-based machine learning, classifier machine learning models, and deep learning approaches. Each method class has its own advantages and disadvantages, so careful consideration is needed to determine the most suitable class when designing a combination therapy prediction method. Future directions to improve current combination therapy prediction technology include incorporation of disease pathobiology, drug characteristics, patient multiomics data, and drug-drug interactions to determine maximally efficacious and tolerable drug regimens for cancer. As computational methods improve in their capability to integrate patient, drug, and disease data, more comprehensive models can be developed to more accurately predict safe and efficacious combination drug therapies for cancer and other complex diseases.
Collapse
Affiliation(s)
- Victoria L. Flanary
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jennifer L. Fisher
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Elizabeth J. Wilk
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Timothy C. Howton
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Brittany N. Lasseigne
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
27
|
Chen J, Wu L, Liu K, Xu Y, He S, Bo X. EDST: a decision stump based ensemble algorithm for synergistic drug combination prediction. BMC Bioinformatics 2023; 24:325. [PMID: 37644423 PMCID: PMC10466832 DOI: 10.1186/s12859-023-05453-3] [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: 06/13/2023] [Accepted: 08/23/2023] [Indexed: 08/31/2023] Open
Abstract
INTRODUCTION There are countless possibilities for drug combinations, which makes it expensive and time-consuming to rely solely on clinical trials to determine the effects of each possible drug combination. In order to screen out the most effective drug combinations more quickly, scholars began to apply machine learning to drug combination prediction. However, most of them are of low interpretability. Consequently, even though they can sometimes produce high prediction accuracy, experts in the medical and biological fields can still not fully rely on their judgments because of the lack of knowledge about the decision-making process. RELATED WORK Decision trees and their ensemble algorithms are considered to be suitable methods for pharmaceutical applications due to their excellent performance and good interpretability. We review existing decision trees or decision tree ensemble algorithms in the medical field and point out their shortcomings. METHOD This study proposes a decision stump (DS)-based solution to extract interpretable knowledge from data sets. In this method, a set of DSs is first generated to selectively form a decision tree (DST). Different from the traditional decision tree, our algorithm not only enables a partial exchange of information between base classifiers by introducing a stump exchange method but also uses a modified Gini index to evaluate stump performance so that the generation of each node is evaluated by a global view to maintain high generalization ability. Furthermore, these trees are combined to construct an ensemble of DST (EDST). EXPERIMENT The two-drug combination data sets are collected from two cell lines with three classes (additive, antagonistic and synergistic effects) to test our method. Experimental results show that both our DST and EDST perform better than other methods. Besides, the rules generated by our methods are more compact and more accurate than other rule-based algorithms. Finally, we also analyze the extracted knowledge by the model in the field of bioinformatics. CONCLUSION The novel decision tree ensemble model can effectively predict the effect of drug combination datasets and easily obtain the decision-making process.
Collapse
Affiliation(s)
| | | | | | - Yong Xu
- Fujian University of Technology, Fuzhou, China
| | - Song He
- Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Xiaochen Bo
- Institute of Health Service and Transfusion Medicine, Beijing, China
| |
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
|
Zhang H, Ek CH, Rattray M, Milo M. SynBa: improved estimation of drug combination synergies with uncertainty quantification. Bioinformatics 2023; 39:i121-i130. [PMID: 37387161 PMCID: PMC10311304 DOI: 10.1093/bioinformatics/btad240] [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] [Indexed: 07/01/2023] Open
Abstract
MOTIVATION There exists a range of different quantification frameworks to estimate the synergistic effect of drug combinations. The diversity and disagreement in estimates make it challenging to determine which combinations from a large drug screening should be proceeded with. Furthermore, the lack of accurate uncertainty quantification for those estimates precludes the choice of optimal drug combinations based on the most favourable synergistic effect. RESULTS In this work, we propose SynBa, a flexible Bayesian approach to estimate the uncertainty of the synergistic efficacy and potency of drug combinations, so that actionable decisions can be derived from the model outputs. The actionability is enabled by incorporating the Hill equation into SynBa, so that the parameters representing the potency and the efficacy can be preserved. Existing knowledge may be conveniently inserted due to the flexibility of the prior, as shown by the empirical Beta prior defined for the normalized maximal inhibition. Through experiments on large combination screenings and comparison against benchmark methods, we show that SynBa provides improved accuracy of dose-response predictions and better-calibrated uncertainty estimation for the parameters and the predictions. AVAILABILITY AND IMPLEMENTATION The code for SynBa is available at https://github.com/HaotingZhang1/SynBa. The datasets are publicly available (DOI of DREAM: 10.7303/syn4231880; DOI of the NCI-ALMANAC subset: 10.5281/zenodo.4135059).
Collapse
Affiliation(s)
- Haoting Zhang
- Department of Computer Science and Technology, University of Cambridge, Cambridge CB3 0FD, United Kingdom
- Health Data Research UK, London NW1 2BE, United Kingdom
| | - Carl Henrik Ek
- Department of Computer Science and Technology, University of Cambridge, Cambridge CB3 0FD, United Kingdom
| | - Magnus Rattray
- Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester M13 9PL, United Kingdom
- Alan Turing Institute, London NW1 2DB, United Kingdom
| | - Marta Milo
- Oncology Data Science, Oncology R&D AstraZeneca, Cambridge CB2 8PA, United Kingdom
| |
Collapse
|
30
|
Srivathsa AV, Sadashivappa NM, Hegde AK, Radha S, Mahesh AR, Ammunje DN, Sen D, Theivendren P, Govindaraj S, Kunjiappan S, Pavadai P. A Review on Artificial Intelligence Approaches and Rational Approaches in Drug Discovery. Curr Pharm Des 2023; 29:1180-1192. [PMID: 37132148 DOI: 10.2174/1381612829666230428110542] [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: 11/30/2022] [Revised: 02/06/2023] [Accepted: 02/27/2023] [Indexed: 05/04/2023]
Abstract
Artificial intelligence (AI) speeds up the drug development process and reduces its time, as well as the cost which is of enormous importance in outbreaks such as COVID-19. It uses a set of machine learning algorithms that collects the available data from resources, categorises, processes and develops novel learning methodologies. Virtual screening is a successful application of AI, which is used in screening huge drug-like databases and filtering to a small number of compounds. The brain's thinking of AI is its neural networking which uses techniques such as Convoluted Neural Network (CNN), Recursive Neural Network (RNN) or Generative Adversial Neural Network (GANN). The application ranges from small molecule drug discovery to the development of vaccines. In the present review article, we discussed various techniques of drug design, structure and ligand-based, pharmacokinetics and toxicity prediction using AI. The rapid phase of discovery is the need of the hour and AI is a targeted approach to achieve this.
Collapse
Affiliation(s)
- Anjana Vidya Srivathsa
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, M.S.R. Nagar, Bengaluru, 560054, India
| | - Nandini Markuli Sadashivappa
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, M.S.R. Nagar, Bengaluru, 560054, India
| | - Apeksha Krishnamurthy Hegde
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, M.S.R. Nagar, Bengaluru, 560054, India
| | - Srimathi Radha
- Department of Pharmaceutical Chemistry, SRM College of Pharmacy, Faculty of Medicine and Health Sciences, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur, Tamil Nadu, 603203, India
| | - Agasa Ramu Mahesh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, M.S.R. Nagar, Bengaluru, 560054, India
| | - Damodar Nayak Ammunje
- Department of Pharmacology, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, M.S.R. Nagar, Bengaluru, 560054, India
| | - Debanjan Sen
- Department of Pharmaceutical Chemistry, BCDA College of Pharmacy & Technology, Hridaypur, Kolkata, 700127, West Bengal, India
| | - Panneerselvam Theivendren
- Department of Pharmaceutical Chemistry, Swamy Vivekanandha College of Pharmacy, Elayampalayam, Tiruchengode, 637205, India
| | - Saravanan Govindaraj
- Department of Pharmaceutical Chemistry, MNR College of Pharmacy, Fasalwadi, Sangareddy, 502 001, India
| | - Selvaraj Kunjiappan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, 626126, India
| | - Parasuraman Pavadai
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, M.S.R. Nagar, Bengaluru, 560054, India
| |
Collapse
|
31
|
Rønneberg L, Kirk PDW, Zucknick M. Dose-response prediction for in-vitro drug combination datasets: a probabilistic approach. BMC Bioinformatics 2023; 24:161. [PMID: 37085771 PMCID: PMC10120211 DOI: 10.1186/s12859-023-05256-6] [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: 10/14/2022] [Accepted: 03/28/2023] [Indexed: 04/23/2023] Open
Abstract
In this paper we propose PIICM, a probabilistic framework for dose-response prediction in high-throughput drug combination datasets. PIICM utilizes a permutation invariant version of the intrinsic co-regionalization model for multi-output Gaussian process regression, to predict dose-response surfaces in untested drug combination experiments. Coupled with an observation model that incorporates experimental uncertainty, PIICM is able to learn from noisily observed cell-viability measurements in settings where the underlying dose-response experiments are of varying quality, utilize different experimental designs, and the resulting training dataset is sparsely observed. We show that the model can accurately predict dose-response in held out experiments, and the resulting function captures relevant features indicating synergistic interaction between drugs.
Collapse
Affiliation(s)
- Leiv Rønneberg
- Oslo Centre for Biostatistics and Epidemiology, University of Oslo, Oslo, Norway
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Paul D W Kirk
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
- Ovarian Cancer Programme, Cancer Research UK Cambridge Centre, Cambridge, UK
| | - Manuela Zucknick
- Oslo Centre for Biostatistics and Epidemiology, University of Oslo, Oslo, Norway.
| |
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
|
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
|
34
|
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
|
35
|
Johanssen T, McVeigh L, Erridge S, Higgins G, Straehla J, Frame M, Aittokallio T, Carragher NO, Ebner D. Glioblastoma and the search for non-hypothesis driven combination therapeutics in academia. Front Oncol 2023; 12:1075559. [PMID: 36733367 PMCID: PMC9886867 DOI: 10.3389/fonc.2022.1075559] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
Glioblastoma (GBM) remains a cancer of high unmet clinical need. Current standard of care for GBM, consisting of maximal surgical resection, followed by ionisation radiation (IR) plus concomitant and adjuvant temozolomide (TMZ), provides less than 15-month survival benefit. Efforts by conventional drug discovery to improve overall survival have failed to overcome challenges presented by inherent tumor heterogeneity, therapeutic resistance attributed to GBM stem cells, and tumor niches supporting self-renewal. In this review we describe the steps academic researchers are taking to address these limitations in high throughput screening programs to identify novel GBM combinatorial targets. We detail how they are implementing more physiologically relevant phenotypic assays which better recapitulate key areas of disease biology coupled with more focussed libraries of small compounds, such as drug repurposing, target discovery, pharmacologically active and novel, more comprehensive anti-cancer target-annotated compound libraries. Herein, we discuss the rationale for current GBM combination trials and the need for more systematic and transparent strategies for identification, validation and prioritisation of combinations that lead to clinical trials. Finally, we make specific recommendations to the preclinical, small compound screening paradigm that could increase the likelihood of identifying tractable, combinatorial, small molecule inhibitors and better drug targets specific to GBM.
Collapse
Affiliation(s)
- Timothy Johanssen
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Laura McVeigh
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Sara Erridge
- Edinburgh Cancer Centre, Western General Hospital, Edinburgh, United Kingdom
| | - Geoffrey Higgins
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Joelle Straehla
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Division of Pediatric Hematology/Oncology, Boston Children’s Hospital, Boston, MA, United States
| | - Margaret Frame
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway
- Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Neil O. Carragher
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Daniel Ebner
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
36
|
Kumar K, Kumar P, Deb D, Unguresan ML, Muresan V. Artificial Intelligence and Machine Learning Based Intervention in Medical Infrastructure: A Review and Future Trends. Healthcare (Basel) 2023; 11:healthcare11020207. [PMID: 36673575 PMCID: PMC9859198 DOI: 10.3390/healthcare11020207] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/01/2023] [Accepted: 01/04/2023] [Indexed: 01/13/2023] Open
Abstract
People in the life sciences who work with Artificial Intelligence (AI) and Machine Learning (ML) are under increased pressure to develop algorithms faster than ever. The possibility of revealing innovative insights and speeding breakthroughs lies in using large datasets integrated on several levels. However, even if there is more data at our disposal than ever, only a meager portion is being filtered, interpreted, integrated, and analyzed. The subject of this technology is the study of how computers may learn from data and imitate human mental processes. Both an increase in the learning capacity and the provision of a decision support system at a size that is redefining the future of healthcare are enabled by AI and ML. This article offers a survey of the uses of AI and ML in the healthcare industry, with a particular emphasis on clinical, developmental, administrative, and global health implementations to support the healthcare infrastructure as a whole, along with the impact and expectations of each component of healthcare. Additionally, possible future trends and scopes of the utilization of this technology in medical infrastructure have also been discussed.
Collapse
Affiliation(s)
- Kamlesh Kumar
- Department of Electrical and Computer Science Engineering, Institute of Infrastructure Technology Research And Management, Ahmedabad 380026, India
| | - Prince Kumar
- Department of Electrical and Computer Science Engineering, Institute of Infrastructure Technology Research And Management, Ahmedabad 380026, India
| | - Dipankar Deb
- Department of Electrical and Computer Science Engineering, Institute of Infrastructure Technology Research And Management, Ahmedabad 380026, India
- Correspondence:
| | | | - Vlad Muresan
- Department of Automation, Technical University of Cluj-Napoca, 400114 Cluj-Napoca, Romania
| |
Collapse
|
37
|
Um SH, Seo Y, Seo H, Lee K, Park SH, Jeon JH, Lim JY, Ok MR, Kim YC, Kim H, Cheon CH, Han HS, Edwards JR, Kim SW, Jeon H. Biomimetic hydrogel blanket for conserving and recovering intrinsic cell properties. Biomater Res 2022; 26:78. [PMID: 36514131 PMCID: PMC9746181 DOI: 10.1186/s40824-022-00327-w] [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: 07/04/2022] [Accepted: 11/20/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cells in the human body experience different growth environments and conditions, such as compressive pressure and oxygen concentrations, depending on the type and location of the tissue. Thus, a culture device that emulates the environment inside the body is required to study cells outside the body. METHODS A blanket-type cell culture device (Direct Contact Pressing: DCP) was fabricated with an alginate-based hydrogel. Changes in cell morphology due to DCP pressure were observed using a phase contrast microscope. The changes in the oxygen permeability and pressure according to the hydrogel concentration of DCP were analyzed. To compare the effects of DCP with normal or artificial hypoxic cultures, cells were divided based on the culture technique: normal culture, DCP culture device, and artificial hypoxic environment. Changes in phenotype, genes, and glycosaminoglycan amounts according to each environment were evaluated. Based on this, the mechanism of each culture environment on the intrinsic properties of conserving chondrocytes was suggested. RESULTS Chondrocytes live under pressure from the surrounding collagen tissue and experience a hypoxic environment because collagen inhibits oxygen permeability. By culturing the chondrocytes in a DCP environment, the capability of DCP to produce a low-oxygen and physical pressure environment was verified. When human primary chondrocytes, which require pressure and a low-oxygen environment during culture to maintain their innate properties, were cultured using the hydrogel blanket, the original shapes and properties of the chondrocytes were maintained. The intrinsic properties could be recovered even in aged cells that had lost their original cell properties. CONCLUSIONS A DCP culture method using a biomimetic hydrogel blanket provides cells with an adjustable physical pressure and a low-oxygen environment. Through this technique, we could maintain the original cellular phenotypes and intrinsic properties of human primary chondrocytes. The results of this study can be applied to other cells that require special pressure and oxygen concentration control to maintain their intrinsic properties. Additionally, this technique has the potential to be applied to the re-differentiation of cells that have lost their original properties.
Collapse
Affiliation(s)
- Seung-Hoon Um
- grid.35541.360000000121053345Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), 02792 Seoul, Republic of Korea ,grid.23856.3a0000 0004 1936 8390Laboratory for Biomaterials and Bioengineering, Department of Min-Met-Materials Engineering, Research Center of CHU de Quebec, Division of Regenerative Medicine, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Laval University, G1V 0A6 Quebec City, Quebec, Canada
| | - Youngmin Seo
- grid.35541.360000000121053345Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), 02792 Seoul, Republic of Korea ,R&D Institute, OID Ltd, Seoul, 06286 Republic of Korea
| | - Hyunseon Seo
- grid.35541.360000000121053345Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), 02792 Seoul, Republic of Korea ,grid.264381.a0000 0001 2181 989XSchool of Medicine, Sungkyunkwan University, Suwon, 16419 Republic of Korea
| | - Kyungwoo Lee
- grid.35541.360000000121053345Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), 02792 Seoul, Republic of Korea
| | - Sun Hwa Park
- grid.23856.3a0000 0004 1936 8390Laboratory for Biomaterials and Bioengineering, Department of Min-Met-Materials Engineering, Research Center of CHU de Quebec, Division of Regenerative Medicine, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Laval University, G1V 0A6 Quebec City, Quebec, Canada
| | - Jung Ho Jeon
- grid.411947.e0000 0004 0470 4224Department of Otolaryngology-Head and Neck Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea ,grid.411947.e0000 0004 0470 4224Department of Biomedicine & Health Sciences, Department of Otolaryngology-Head and Neck Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Yeon Lim
- grid.411947.e0000 0004 0470 4224Department of Otolaryngology-Head and Neck Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Myoung-Ryul Ok
- grid.35541.360000000121053345Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), 02792 Seoul, Republic of Korea
| | - Yu-Chan Kim
- grid.35541.360000000121053345Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), 02792 Seoul, Republic of Korea ,grid.412786.e0000 0004 1791 8264Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792 Republic of Korea
| | - Hyunjung Kim
- grid.256753.00000 0004 0470 5964Division of Nursing, Research Institute of Nursing Science, Hallym University, Chuncheon, 24252 Republic of Korea
| | - Cheol-Hong Cheon
- grid.222754.40000 0001 0840 2678Department of Chemistry, Korea University, Seoul, 02841 Republic of Korea
| | - Hyung-Seop Han
- grid.35541.360000000121053345Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), 02792 Seoul, Republic of Korea
| | - James R. Edwards
- grid.4991.50000 0004 1936 8948Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), Botnar Research Centre, University of Oxford, Oxford, OX3 7LD UK
| | - Sung Won Kim
- grid.411947.e0000 0004 0470 4224Department of Otolaryngology-Head and Neck Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea ,grid.411947.e0000 0004 0470 4224Department of Biomedicine & Health Sciences, Department of Otolaryngology-Head and Neck Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hojeong Jeon
- grid.35541.360000000121053345Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), 02792 Seoul, Republic of Korea ,grid.412786.e0000 0004 1791 8264Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792 Republic of Korea ,grid.222754.40000 0001 0840 2678KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841 Republic of Korea
| |
Collapse
|
38
|
Askr H, Elgeldawi E, Aboul Ella H, Elshaier YAMM, Gomaa MM, Hassanien AE. Deep learning in drug discovery: an integrative review and future challenges. Artif Intell Rev 2022; 56:5975-6037. [PMID: 36415536 PMCID: PMC9669545 DOI: 10.1007/s10462-022-10306-1] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2022] [Indexed: 11/18/2022]
Abstract
Recently, using artificial intelligence (AI) in drug discovery has received much attention since it significantly shortens the time and cost of developing new drugs. Deep learning (DL)-based approaches are increasingly being used in all stages of drug development as DL technology advances, and drug-related data grows. Therefore, this paper presents a systematic Literature review (SLR) that integrates the recent DL technologies and applications in drug discovery Including, drug-target interactions (DTIs), drug-drug similarity interactions (DDIs), drug sensitivity and responsiveness, and drug-side effect predictions. We present a review of more than 300 articles between 2000 and 2022. The benchmark data sets, the databases, and the evaluation measures are also presented. In addition, this paper provides an overview of how explainable AI (XAI) supports drug discovery problems. The drug dosing optimization and success stories are discussed as well. Finally, digital twining (DT) and open issues are suggested as future research challenges for drug discovery problems. Challenges to be addressed, future research directions are identified, and an extensive bibliography is also included.
Collapse
Affiliation(s)
- Heba Askr
- Faculty of Computers and Artificial Intelligence, University of Sadat City, Sadat City, Egypt
| | - Enas Elgeldawi
- Computer Science Department, Faculty of Science, Minia University, Minia, Egypt
| | - Heba Aboul Ella
- Faculty of Pharmacy and Drug Technology, Chinese University in Egypt (CUE), Cairo, Egypt
| | | | - Mamdouh M. Gomaa
- Computer Science Department, Faculty of Science, Minia University, Minia, Egypt
| | - Aboul Ella Hassanien
- Faculty of Computers and Artificial Intelligence, Cairo University, Cairo, Egypt
| |
Collapse
|
39
|
Larkins-Ford J, Degefu YN, Van N, Sokolov A, Aldridge BB. Design principles to assemble drug combinations for effective tuberculosis therapy using interpretable pairwise drug response measurements. Cell Rep Med 2022; 3:100737. [PMID: 36084643 PMCID: PMC9512659 DOI: 10.1016/j.xcrm.2022.100737] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 05/16/2022] [Accepted: 08/16/2022] [Indexed: 11/02/2022]
Abstract
A challenge in tuberculosis treatment regimen design is the necessity to combine three or more antibiotics. We narrow the prohibitively large search space by breaking down high-order drug combinations into drug pair units. Using pairwise in vitro measurements, we train machine learning models to predict higher-order combination treatment outcomes in the relapsing BALB/c mouse model. Classifiers perform well and predict many of the >500 possible combinations among 12 antibiotics to be improved over bedaquiline + pretomanid + linezolid, a treatment-shortening regimen compared with the standard of care in mice. We reformulate classifiers as simple rulesets to reveal guiding principles of constructing combination therapies for both preclinical and clinical outcomes. One example ruleset combines a drug pair that is synergistic in a dormancy model with a pair that is potent in a cholesterol-rich growth environment. These rulesets are predictive, intuitive, and practical, thus enabling rational construction of drug combinations. Evaluate the large drug combination space for potential tuberculosis treatments In vitro 2-drug combination measurements predict 3–4 drug treatment outcomes in vivo Strongly synergistic, antagonistic, or potent drug pairs drive treatment outcome Simple rules articulate drug combination design principles for tuberculosis
Collapse
|
40
|
Liu X, Song C, Liu S, Li M, Zhou X, Zhang W. Multi-way relation-enhanced hypergraph representation learning for anti-cancer drug synergy prediction. Bioinformatics 2022; 38:4782-4789. [PMID: 36000898 DOI: 10.1093/bioinformatics/btac579] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/14/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
MOTIVATION Drug combinations have exhibited promise in treating cancers with less toxicity and fewer adverse reactions. However, in vitro screening of synergistic drug combinations is time-consuming and labour-intensive because of the combinatorial explosion. Although a number of computational methods have been developed for predicting synergistic drug combinations, the multi-way relations between drug combinations and cell lines existing in drug synergy data have not been well exploited. RESULTS We propose a multi-way relation-enhanced hypergraph representation learning method to predict anti-cancer drug synergy, named HypergraphSynergy. HypergraphSynergy formulates synergistic drug combinations over cancer cell lines as a hypergraph, in which drugs and cell lines are represented by nodes and synergistic drug-drug-cell line triplets are represented by hyperedges, and leverages the biochemical features of drugs and cell lines as node attributes. Then, a hypergraph neural network is designed to learn the embeddings of drugs and cell lines from the hypergraph and predict drug synergy. Moreover, the auxiliary task of reconstructing the similarity networks of drugs and cell lines is considered to enhance the generalization ability of the model. In the computational experiments, HypergraphSynergy outperforms other state-of-the-art synergy prediction methods on two benchmark datasets for both classification and regression tasks, and is applicable to unseen drug combinations or cell lines. The studies revealed that the hypergraph formulation allows us to capture and explain complex multi-way relations of drug combinations and cell lines, and also provides a flexible framework to make the best use of diverse information. AVAILABILITY AND IMPLEMENTATION The source data and codes of HypergraphSynergy can be freely downloaded from https://github.com/liuxuan666/HypergraphSynergy. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Xuan Liu
- College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Congzhi Song
- College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Shichao Liu
- College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Menglu Li
- College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xionghui Zhou
- College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wen Zhang
- College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China.,Agricultural Bioinformatics Key Laboratory of Hubei Province, Hubei Engineering Technology Research Center of Agricultural Big Data, Key Laboratory of Smart Animal Farming Technology, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, 430070, China
| |
Collapse
|
41
|
Ding P, Pan Y, Wang Q, Xu R. Prediction and evaluation of combination pharmacotherapy using natural language processing, machine learning and patient electronic health records. J Biomed Inform 2022; 133:104164. [PMID: 35985621 DOI: 10.1016/j.jbi.2022.104164] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/18/2022]
Abstract
Combination pharmacotherapy targets key disease pathways in a synergistic or additive manner and has high potential in treating complex diseases. Computational methods have been developed to identifying combination pharmacotherapy by analyzing large amounts of biomedical data. Existing computational approaches are often underpowered due to their reliance on our limited understanding of disease mechanisms. On the other hand, observable phenotypic inter-relationships among thousands of diseases often reflect their underlying shared genetic and molecular underpinnings, therefore can offer unique opportunities to design computational models to discover novel combinational therapies by automatically transferring knowledge among phenotypically related diseases. We developed a novel phenome-driven drug discovery system, named TuSDC, which leverages knowledge of existing drug combinations, disease comorbidities, and disease treatments of thousands of disease and drug entities extracted from over 31.5 million biomedical research articles using natural language processing techniques. TuSDC predicts combination pharmacotherapy by extracting representations of diseases and drugs using tensor factorization approaches. In external validation, TuSDC achieved an average precision of 0.77 for top ranked candidates, outperforming a state of art mechanism-based method for discovering drug combinations in treating hypertension. We evaluated top ranked anti-hypertension drug combinations using electronic health records of 84.7 million unique patients and showed that a novel drug combination hydrochlorothiazide-digoxin was associated with significantly lower hazards of subsequent hypertension as compared to the monotherapy hydrochlorothiazide alone (HR: 0.769, 95% CI [0.732, 0.807]) and digoxin alone (0.857, 95% CI [0.785, 0.936]). Data-driven informatics analyses reveal that the renin-angiotensin system is involved in the synergistical interactions of hydrochlorothiazide and digoxin on regulating hypertension. The prediction model's code with PyTorch version 1.5 is available at http://nlp.case.edu/public/data/TuSDC/.
Collapse
Affiliation(s)
- Pingjian Ding
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Yiheng Pan
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Quanqiu Wang
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Rong Xu
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
42
|
Lv J, Liu G, Hao J, Ju Y, Sun B, Sun Y. Computational models, databases and tools for antibiotic combinations. Brief Bioinform 2022; 23:6652783. [PMID: 35915052 DOI: 10.1093/bib/bbac309] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Antibiotic combination is a promising strategy to extend the lifetime of antibiotics and thereby combat antimicrobial resistance. However, screening for new antibiotic combinations is both time-consuming and labor-intensive. In recent years, an increasing number of researchers have used computational models to predict effective antibiotic combinations. In this review, we summarized existing computational models for antibiotic combinations and discussed the limitations and challenges of these models in detail. In addition, we also collected and summarized available data resources and tools for antibiotic combinations. This study aims to help computational biologists design more accurate and interpretable computational models.
Collapse
Affiliation(s)
- Ji Lv
- College of Computer Science and Technology, Jilin University, Changchun, China.,Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, China
| | - Guixia Liu
- College of Computer Science and Technology, Jilin University, Changchun, China.,Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, China
| | - Junli Hao
- College of Food Science, Northeast Agricultural University, Harbin, China
| | - Yuan Ju
- Sichuan University Library, Sichuan University, Chengdu, China
| | - Binwen Sun
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumor Therapy, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ying Sun
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, China
| |
Collapse
|
43
|
Rintala TJ, Ghosh A, Fortino V. Network approaches for modeling the effect of drugs and diseases. Brief Bioinform 2022; 23:6608969. [PMID: 35704883 PMCID: PMC9294412 DOI: 10.1093/bib/bbac229] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/29/2022] [Accepted: 05/17/2021] [Indexed: 12/12/2022] Open
Abstract
The network approach is quickly becoming a fundamental building block of computational methods aiming at elucidating the mechanism of action (MoA) and therapeutic effect of drugs. By modeling the effect of drugs and diseases on different biological networks, it is possible to better explain the interplay between disease perturbations and drug targets as well as how drug compounds induce favorable biological responses and/or adverse effects. Omics technologies have been extensively used to generate the data needed to study the mechanisms of action of drugs and diseases. These data are often exploited to define condition-specific networks and to study whether drugs can reverse disease perturbations. In this review, we describe network data mining algorithms that are commonly used to study drug’s MoA and to improve our understanding of the basis of chronic diseases. These methods can support fundamental stages of the drug development process, including the identification of putative drug targets, the in silico screening of drug compounds and drug combinations for the treatment of diseases. We also discuss recent studies using biological and omics-driven networks to search for possible repurposed FDA-approved drug treatments for SARS-CoV-2 infections (COVID-19).
Collapse
Affiliation(s)
- T J Rintala
- Institute of Biomedicine, University of Eastern Finland, 70210 Kuopio, Finland
| | - Arindam Ghosh
- Institute of Biomedicine, University of Eastern Finland, 70210 Kuopio, Finland
| | - V Fortino
- Institute of Biomedicine, University of Eastern Finland, 70210 Kuopio, Finland
| |
Collapse
|
44
|
Jafari M, Mirzaie M, Bao J, Barneh F, Zheng S, Eriksson J, Heckman CA, Tang J. Bipartite network models to design combination therapies in acute myeloid leukaemia. Nat Commun 2022; 13:2128. [PMID: 35440130 PMCID: PMC9018865 DOI: 10.1038/s41467-022-29793-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 03/30/2022] [Indexed: 12/20/2022] Open
Abstract
Combination therapy is preferred over single-targeted monotherapies for cancer treatment due to its efficiency and safety. However, identifying effective drug combinations costs time and resources. We propose a method for identifying potential drug combinations by bipartite network modelling of patient-related drug response data, specifically the Beat AML dataset. The median of cell viability is used as a drug potency measurement to reconstruct a weighted bipartite network, model drug-biological sample interactions, and find the clusters of nodes inside two projected networks. Then, the clustering results are leveraged to discover effective multi-targeted drug combinations, which are also supported by more evidence using GDSC and ALMANAC databases. The potency and synergy levels of selective drug combinations are corroborated against monotherapy in three cell lines for acute myeloid leukaemia in vitro. In this study, we introduce a nominal data mining approach to improving acute myeloid leukaemia treatment through combinatorial therapy. Identifying effective drug combinations to treat cancer is a challenging task, either experimentally or computationally. Here, the authors develop a bipartite network modelling approach to propose drug combination strategies in acute myeloid leukaemia using patient and cell line drug screening data.
Collapse
Affiliation(s)
- Mohieddin Jafari
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Mehdi Mirzaie
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jie Bao
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Farnaz Barneh
- Prinses Maxima Center for Pediatric Oncology, 3584 CS Utrecht, Utrech, the Netherlands
| | - Shuyu Zheng
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Johanna Eriksson
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Caroline A Heckman
- Institute for Molecular Medicine Finland - FIMM, HiLIFE - Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Jing Tang
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
45
|
Aittokallio T. What are the current challenges for machine learning in drug discovery and repurposing? Expert Opin Drug Discov 2022; 17:423-425. [PMID: 35255749 DOI: 10.1080/17460441.2022.2050694] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland.,Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway.,Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
46
|
Pancreatic Cancer Organoids in the Field of Precision Medicine: A Review of Literature and Experience on Drug Sensitivity Testing with Multiple Readouts and Synergy Scoring. Cancers (Basel) 2022; 14:cancers14030525. [PMID: 35158794 PMCID: PMC8833348 DOI: 10.3390/cancers14030525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/11/2022] [Accepted: 01/16/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary New treatments are urgently needed for pancreatic ductal adenocarcinoma because it is one of the most aggressive and lethal cancers, detected too late and resistant to conventional chemotherapy. Tumors in most patients feature a similar set of core mutations but so far it has not been possible to design a one-fits-all treatment strategy. Instead, efforts are underway to personalize the therapies. To find the treatments that might work the best for each patient, entirely new experimental platforms based on living miniature tumors, organoids, have been developed. We review here the latest international findings in designing personalized treatments pancreatic cancer patients using organoids as testing beds. Our own work adds important clues about how such testing could, and perhaps should, be conducted. Abstract Pancreatic ductal adenocarcinoma (PDAC) is a silent killer, often diagnosed late. However, it is also dishearteningly resistant to nearly all forms of treatment. New therapies are urgently needed, and with the advent of organoid culture for pancreatic cancer, an increasing number of innovative approaches are being tested. Organoids can be derived within a short enough time window to allow testing of several anticancer agents, which opens up the possibility for functional precision medicine for pancreatic cancer. At the same time, organoid model systems are being refined to better mimic the cancer, for example, by incorporation of components of the tumor microenvironment. We review some of the latest developments in pancreatic cancer organoid research and in novel treatment design. We also summarize our own current experiences with pancreatic cancer organoid drug sensitivity and resistance testing (DSRT) in 14 organoids from 11 PDAC patients. Our data show that it may be necessary to include a cell death read-out in ex vivo DSRT assays, as metabolic viability quantitation does not capture actual organoid killing. We also successfully adapted the organoid platform for drug combination synergy discovery. Lastly, live organoid culture 3D confocal microscopy can help identify individual surviving tumor cells escaping cell death even during harsh combination treatments. Taken together, the organoid technology allows the development of novel precision medicine approaches for PDAC, which paves the way for clinical trials and much needed new treatment options for pancreatic cancer patients.
Collapse
|
47
|
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
|
48
|
Kong W, Midena G, Chen Y, Athanasiadis P, Wang T, Rousu J, He L, Aittokallio T. Systematic review of computational methods for drug combination prediction. Comput Struct Biotechnol J 2022; 20:2807-2814. [PMID: 35685365 PMCID: PMC9168078 DOI: 10.1016/j.csbj.2022.05.055] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 12/26/2022] Open
Abstract
Synergistic effects between drugs are rare and highly context-dependent and patient-specific. Hence, there is a need to develop novel approaches to stratify patients for optimal therapy regimens, especially in the context of personalized design of combinatorial treatments. Computational methods enable systematic in-silico screening of combination effects, and can thereby prioritize most potent combinations for further testing, among the massive number of potential combinations. To help researchers to choose a prediction method that best fits for various real-world applications, we carried out a systematic literature review of 117 computational methods developed to date for drug combination prediction, and classified the methods in terms of their combination prediction tasks and input data requirements. Most current methods focus on prediction or classification of combination synergy, and only a few methods consider the efficacy and potential toxicity of the combinations, which are the key determinants of therapeutic success of drug treatments. Furthermore, there is a need to further develop methods that enable dose-specific predictions of combination effects across multiple doses, which is important for clinical translation of the predictions, as well as model-based identification of biomarkers predictive of heterogeneous drug combination responses. Even if most of the computational methods reviewed focus on anticancer applications, many of the modelling approaches are also applicable to antiviral and other diseases or indications.
Collapse
|
49
|
Athanasiadis P, Ianevski A, Skånland SS, Aittokallio T. Computational Pipeline for Rational Drug Combination Screening in Patient-Derived Cells. Methods Mol Biol 2022; 2449:327-348. [PMID: 35507270 DOI: 10.1007/978-1-0716-2095-3_14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In many complex diseases, such as cancers, resistance to monotherapies easily occurs, and longer-term treatment responses often require combinatorial therapies as next-line regimens. However, due to a massive number of possible drug combinations to test, there is a need for systematic and rational approaches to finding safe and effective drug combinations for each individual patient. This protocol describes an ecosystem of computational methods to guide high-throughput combinatorial screening that help experimental researchers to identify optimal drug combinations in terms of synergy, efficacy, and/or selectivity for further preclinical and clinical investigation. The methods are demonstrated in the context of combinatorial screening in primary cells of leukemia patients, where the translational aim is to identify drug combinations that show not only high synergy but also maximal cancer-selectivity. The mechanism-agnostic and cost-effective computational methods are widely applicable to various cancer types, which are amenable to drug testing, as the computational methods take as input only the phenotypic measurements of a subset of drug combinations, without requiring target information or genomic profiles of the patient samples.
Collapse
Affiliation(s)
- Paschalis Athanasiadis
- Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology, University of Oslo, Oslo, Norway
| | - Aleksandr Ianevski
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Sigrid S Skånland
- Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K. G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tero Aittokallio
- Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
- Oslo Centre for Biostatistics and Epidemiology, University of Oslo, Oslo, Norway.
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
50
|
Network Biology and Artificial Intelligence Drive the Understanding of the Multidrug Resistance Phenotype in Cancer. Drug Resist Updat 2022; 60:100811. [DOI: 10.1016/j.drup.2022.100811] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 02/07/2023]
|