1
|
Azadinejad H, Farhadi Rad M, Shariftabrizi A, Rahmim A, Abdollahi H. Optimizing Cancer Treatment: Exploring the Role of AI in Radioimmunotherapy. Diagnostics (Basel) 2025; 15:397. [PMID: 39941326 PMCID: PMC11816985 DOI: 10.3390/diagnostics15030397] [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: 12/10/2024] [Revised: 01/14/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Radioimmunotherapy (RIT) is a novel cancer treatment that combines radiotherapy and immunotherapy to precisely target tumor antigens using monoclonal antibodies conjugated with radioactive isotopes. This approach offers personalized, systemic, and durable treatment, making it effective in cancers resistant to conventional therapies. Advances in artificial intelligence (AI) present opportunities to enhance RIT by improving precision, efficiency, and personalization. AI plays a critical role in patient selection, treatment planning, dosimetry, and response assessment, while also contributing to drug design and tumor classification. This review explores the integration of AI into RIT, emphasizing its potential to optimize the entire treatment process and advance personalized cancer care.
Collapse
Affiliation(s)
- Hossein Azadinejad
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah 6714869914, Iran;
| | - Mohammad Farhadi Rad
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
| | - Ahmad Shariftabrizi
- Department of Radiology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA;
| | - Arman Rahmim
- Department of Radiology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 0B4, Canada
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Hamid Abdollahi
- Department of Radiology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 0B4, Canada
| |
Collapse
|
2
|
Camilleri F, Wenda JM, Pecoraro-Mercier C, Comet JP, Rouquié D. Cell Painting and Chemical Structure Read-Across Can Complement Each Other for Rat Acute Oral Toxicity Prediction in Chemical Early Derisking. Chem Res Toxicol 2024; 37:1851-1866. [PMID: 39513413 DOI: 10.1021/acs.chemrestox.4c00169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Early derisking decisions in the development of new chemical compounds enable the identification of novel chemical candidates with improved safety profiles. In vivo studies are traditionally conducted in the early assessment of acute oral toxicity of crop protection products to avoid compounds, which are considered "very acutely toxic", with an in vivo lethal dose of 50% (LD50) ≤ 60 mg/kg body weight. Those studies are lengthy and costly and raise ethical concerns, catalyzing the use of nonanimal alternatives. The objective of our analysis was to assess the predictive efficacy of read-across approaches for acute oral toxicity in rats, comparing the use of chemical structure information, in vitro biological data derived from the Cell Painting profiling assay on U2OS cells, or the combination of both. Our findings indicate that the classification of compounds as very acute oral toxic (LD50 ≤ 60 mg/kg) or not is possible using a read-across approach, with chemical structure information, morphological profiles, or a combination of both. When classifying compounds structurally similar to those in the training set, the chemical structure was more predictive (balanced accuracy of 0.82). Conversely, when the compounds to be classified were structurally different from those in the training set, the morphological profiles were more predictive (balanced accuracy of 0.72). Combining the two models allowed for the classification of compounds structurally similar to those in the training set to slightly improve the predictions (balanced accuracy of 0.85).
Collapse
Affiliation(s)
- Fabrice Camilleri
- Toxicology Data Science, Bayer SAS Crop Science Division, 355 rue Dostoïevski, CS 90153 Valbonne, Cedex, Sophia Antipolis 06906, France
- I3S UMR 7271 du CNRS, Université Côte d'Azur, Bâtiment Algorithme-Euclide-B, 2000 Route des Lucioles, B.P. 121, Sophia Antipolis 06903, France
| | - Joanna M Wenda
- Early Toxicology, Bayer SAS Crop Science Division, 355 rue Dostoïevski, CS 90153 Valbonne, Cedex, Sophia Antipolis 06906, France
| | - Claire Pecoraro-Mercier
- Early Toxicology, Bayer SAS Crop Science Division, 355 rue Dostoïevski, CS 90153 Valbonne, Cedex, Sophia Antipolis 06906, France
| | - Jean-Paul Comet
- I3S UMR 7271 du CNRS, Université Côte d'Azur, Bâtiment Algorithme-Euclide-B, 2000 Route des Lucioles, B.P. 121, Sophia Antipolis 06903, France
| | - David Rouquié
- Toxicology Data Science, Bayer SAS Crop Science Division, 355 rue Dostoïevski, CS 90153 Valbonne, Cedex, Sophia Antipolis 06906, France
| |
Collapse
|
3
|
Harrill JA, Everett LJ, Haggard DE, Word LJ, Bundy JL, Chambers B, Harris F, Willis C, Thomas RS, Shah I, Judson R. Signature analysis of high-throughput transcriptomics screening data for mechanistic inference and chemical grouping. Toxicol Sci 2024; 202:103-122. [PMID: 39177380 DOI: 10.1093/toxsci/kfae108] [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] [Indexed: 08/24/2024] Open
Abstract
High-throughput transcriptomics (HTTr) uses gene expression profiling to characterize the biological activity of chemicals in in vitro cell-based test systems. As an extension of a previous study testing 44 chemicals, HTTr was used to screen an additional 1,751 unique chemicals from the EPA's ToxCast collection in MCF7 cells using 8 concentrations and an exposure duration of 6 h. We hypothesized that concentration-response modeling of signature scores could be used to identify putative molecular targets and cluster chemicals with similar bioactivity. Clustering and enrichment analyses were conducted based on signature catalog annotations and ToxPrint chemotypes to facilitate molecular target prediction and grouping of chemicals with similar bioactivity profiles. Enrichment analysis based on signature catalog annotation identified known mechanisms of action (MeOAs) associated with well-studied chemicals and generated putative MeOAs for other active chemicals. Chemicals with predicted MeOAs included those targeting estrogen receptor (ER), glucocorticoid receptor (GR), retinoic acid receptor (RAR), the NRF2/KEAP/ARE pathway, AP-1 activation, and others. Using reference chemicals for ER modulation, the study demonstrated that HTTr in MCF7 cells was able to stratify chemicals in terms of agonist potency, distinguish ER agonists from antagonists, and cluster chemicals with similar activities as predicted by the ToxCast ER Pathway model. Uniform manifold approximation and projection (UMAP) embedding of signature-level results identified novel ER modulators with no ToxCast ER Pathway model predictions. Finally, UMAP combined with ToxPrint chemotype enrichment was used to explore the biological activity of structurally related chemicals. The study demonstrates that HTTr can be used to inform chemical risk assessment by determining in vitro points of departure, predicting chemicals' MeOA and grouping chemicals with similar bioactivity profiles.
Collapse
Affiliation(s)
- Joshua A Harrill
- Center for Computational Toxicology & Exposure, Office of Research and Development, US Environmental Protection Agency, Durham, NC 27711, United States
| | - Logan J Everett
- Center for Computational Toxicology & Exposure, Office of Research and Development, US Environmental Protection Agency, Durham, NC 27711, United States
| | - Derik E Haggard
- Center for Computational Toxicology & Exposure, Office of Research and Development, US Environmental Protection Agency, Durham, NC 27711, United States
| | - Laura J Word
- Center for Computational Toxicology & Exposure, Office of Research and Development, US Environmental Protection Agency, Durham, NC 27711, United States
| | - Joseph L Bundy
- Center for Computational Toxicology & Exposure, Office of Research and Development, US Environmental Protection Agency, Durham, NC 27711, United States
| | - Bryant Chambers
- Center for Computational Toxicology & Exposure, Office of Research and Development, US Environmental Protection Agency, Durham, NC 27711, United States
| | - Felix Harris
- Center for Computational Toxicology & Exposure, Office of Research and Development, US Environmental Protection Agency, Durham, NC 27711, United States
- Oak Ridge Associated Universities (ORAU) National Student Services Contractor, Oak Ridge, TN 37831, United States
| | - Clinton Willis
- Center for Computational Toxicology & Exposure, Office of Research and Development, US Environmental Protection Agency, Durham, NC 27711, United States
| | - Russell S Thomas
- Center for Computational Toxicology & Exposure, Office of Research and Development, US Environmental Protection Agency, Durham, NC 27711, United States
| | - Imran Shah
- Center for Computational Toxicology & Exposure, Office of Research and Development, US Environmental Protection Agency, Durham, NC 27711, United States
| | - Richard Judson
- Center for Computational Toxicology & Exposure, Office of Research and Development, US Environmental Protection Agency, Durham, NC 27711, United States
| |
Collapse
|
4
|
Vinik Y, Maimon A, Raj H, Dubey V, Geist F, Wienke D, Lev S. Computational pipeline predicting cell death suppressors as targets for cancer therapy. iScience 2024; 27:110859. [PMID: 39310772 PMCID: PMC11416655 DOI: 10.1016/j.isci.2024.110859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/24/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Identification of promising targets for cancer therapy is a global effort in precision medicine. Here, we describe a computational pipeline integrating transcriptomic and vulnerability responses to cell-death inducing drugs, to predict cell-death suppressors as candidate targets for cancer therapy. The prediction is based on two modules; the transcriptomic similarity module to identify genes whose targeting results in similar transcriptomic responses of the death-inducing drugs, and the correlation module to identify candidate genes whose expression correlates to the vulnerability of cancer cells to the same death-inducers. The combined predictors of these two modules were integrated into a single metric. As a proof-of-concept, we selected ferroptosis inducers as death-inducing drugs in triple negative breast cancer. The pipeline reliably predicted candidate genes as ferroptosis suppressors, as validated by computational methods and cellular assays. The described pipeline might be used to identify repressors of various cell-death pathways as potential therapeutic targets for different cancer types.
Collapse
Affiliation(s)
- Yaron Vinik
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Avi Maimon
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Harsha Raj
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Vinay Dubey
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Felix Geist
- The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Dirk Wienke
- The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Sima Lev
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
5
|
Fredin Haslum J, Lardeau CH, Karlsson J, Turkki R, Leuchowius KJ, Smith K, Müllers E. Cell Painting-based bioactivity prediction boosts high-throughput screening hit-rates and compound diversity. Nat Commun 2024; 15:3470. [PMID: 38658534 PMCID: PMC11043326 DOI: 10.1038/s41467-024-47171-1] [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: 04/03/2023] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
Identifying active compounds for a target is a time- and resource-intensive task in early drug discovery. Accurate bioactivity prediction using morphological profiles could streamline the process, enabling smaller, more focused compound screens. We investigate the potential of deep learning on unrefined single-concentration activity readouts and Cell Painting data, to predict compound activity across 140 diverse assays. We observe an average ROC-AUC of 0.744 ± 0.108 with 62% of assays achieving ≥0.7, 30% ≥0.8, and 7% ≥0.9. In many cases, the high prediction performance can be achieved using only brightfield images instead of multichannel fluorescence images. A comprehensive analysis shows that Cell Painting-based bioactivity prediction is robust across assay types, technologies, and target classes, with cell-based assays and kinase targets being particularly well-suited for prediction. Experimental validation confirms the enrichment of active compounds. Our findings indicate that models trained on Cell Painting data, combined with a small set of single-concentration data points, can reliably predict the activity of a compound library across diverse targets and assays while maintaining high hit rates and scaffold diversity. This approach has the potential to reduce the size of screening campaigns, saving time and resources, and enabling primary screening with more complex assays.
Collapse
Affiliation(s)
- Johan Fredin Haslum
- KTH Royal Institute of Technology, Stockholm, Sweden
- Science for Life Laboratory, Stockholm, Sweden
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Johan Karlsson
- Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Riku Turkki
- Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Kevin Smith
- KTH Royal Institute of Technology, Stockholm, Sweden
- Science for Life Laboratory, Stockholm, Sweden
| | - Erik Müllers
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| |
Collapse
|
6
|
Godinez WJ, Trifonov V, Fang B, Kuzu G, Pei L, Guiguemde WA, Martin EJ, King FJ, Jenkins JL, Skewes-Cox P. Compound Activity Prediction with Dose-Dependent Transcriptomic Profiles and Deep Learning. J Chem Inf Model 2024; 64:2695-2704. [PMID: 38293736 DOI: 10.1021/acs.jcim.3c01855] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Predicting compound activity in assays is a long-standing challenge in drug discovery. Computational models based on compound-induced gene expression signatures from a single profiling assay have shown promise toward predicting compound activity in other, seemingly unrelated, assays. Applications of such models include predicting mechanisms-of-action (MoA) for phenotypic hits, identifying off-target activities, and identifying polypharmacologies. Here, we introduce transcriptomics-to-activity transformer (TAT) models that leverage gene expression profiles observed over compound treatment at multiple concentrations to predict the compound activity in other biochemical or cellular assays. We built TAT models based on gene expression data from a RASL-seq assay to predict the activity of 2692 compounds in 262 dose-response assays. We obtained useful models for 51% of the assays, as determined through a realistic held-out set. Prospectively, we experimentally validated the activity predictions of a TAT model in a malaria inhibition assay. With a 63% hit rate, TAT successfully identified several submicromolar malaria inhibitors. Our results thus demonstrate the potential of transcriptomic responses over compound concentration and the TAT modeling framework as a cost-efficient way to identify the bioactivities of promising compounds across many assays.
Collapse
Affiliation(s)
- William J Godinez
- Novartis Institutes for BioMedical Research, Emeryville, California 94608, United States
| | - Vladimir Trifonov
- Novartis Institutes for BioMedical Research, San Diego, California 92121, United States
| | - Bin Fang
- Novartis Institutes for BioMedical Research, San Diego, California 92121, United States
| | - Guray Kuzu
- Novartis Institutes for BioMedical Research, San Diego, California 92121, United States
| | - Luying Pei
- Novartis Institutes for BioMedical Research, Emeryville, California 94608, United States
| | - W Armand Guiguemde
- Novartis Institutes for BioMedical Research, Emeryville, California 94608, United States
| | - Eric J Martin
- Novartis Institutes for BioMedical Research, Emeryville, California 94608, United States
| | - Frederick J King
- Novartis Institutes for BioMedical Research, San Diego, California 92121, United States
| | - Jeremy L Jenkins
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Peter Skewes-Cox
- Novartis Institutes for BioMedical Research, Emeryville, California 94608, United States
| |
Collapse
|
7
|
Hosseini-Gerami L, Higgins IA, Collier DA, Laing E, Evans D, Broughton H, Bender A. Benchmarking causal reasoning algorithms for gene expression-based compound mechanism of action analysis. BMC Bioinformatics 2023; 24:154. [PMID: 37072707 PMCID: PMC10111792 DOI: 10.1186/s12859-023-05277-1] [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: 01/07/2022] [Accepted: 04/06/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND Elucidating compound mechanism of action (MoA) is beneficial to drug discovery, but in practice often represents a significant challenge. Causal Reasoning approaches aim to address this situation by inferring dysregulated signalling proteins using transcriptomics data and biological networks; however, a comprehensive benchmarking of such approaches has not yet been reported. Here we benchmarked four causal reasoning algorithms (SigNet, CausalR, CausalR ScanR and CARNIVAL) with four networks (the smaller Omnipath network vs. 3 larger MetaBase™ networks), using LINCS L1000 and CMap microarray data, and assessed to what extent each factor dictated the successful recovery of direct targets and compound-associated signalling pathways in a benchmark dataset comprising 269 compounds. We additionally examined impact on performance in terms of the functions and roles of protein targets and their connectivity bias in the prior knowledge networks. RESULTS According to statistical analysis (negative binomial model), the combination of algorithm and network most significantly dictated the performance of causal reasoning algorithms, with the SigNet recovering the greatest number of direct targets. With respect to the recovery of signalling pathways, CARNIVAL with the Omnipath network was able to recover the most informative pathways containing compound targets, based on the Reactome pathway hierarchy. Additionally, CARNIVAL, SigNet and CausalR ScanR all outperformed baseline gene expression pathway enrichment results. We found no significant difference in performance between L1000 data or microarray data, even when limited to just 978 'landmark' genes. Notably, all causal reasoning algorithms also outperformed pathway recovery based on input DEGs, despite these often being used for pathway enrichment. Causal reasoning methods performance was somewhat correlated with connectivity and biological role of the targets. CONCLUSIONS Overall, we conclude that causal reasoning performs well at recovering signalling proteins related to compound MoA upstream from gene expression changes by leveraging prior knowledge networks, and that the choice of network and algorithm has a profound impact on the performance of causal reasoning algorithms. Based on the analyses presented here this is true for both microarray-based gene expression data as well as those based on the L1000 platform.
Collapse
Affiliation(s)
- Layla Hosseini-Gerami
- Department of Chemistry, Centre for Molecular Informatics, Cambridge, UK
- Ignota Labs, London, UK
| | | | - David A Collier
- Eli Lilly and Company, Bracknell, UK
- Social, Genetic and Developmental Psychiatry Centre, IoPPN, Kings's College London, London, UK
- Genetic and Genomic Consulting Ltd, Farnham, UK
| | - Emma Laing
- Eli Lilly and Company, Bracknell, UK
- GSK, Stevenage, UK
| | - David Evans
- Eli Lilly and Company, Bracknell, UK
- DeepMind, London, UK
| | - Howard Broughton
- Centre de Investigación, Eli Lilly and Company, Alcobendas, Spain
| | - Andreas Bender
- Department of Chemistry, Centre for Molecular Informatics, Cambridge, UK.
| |
Collapse
|
8
|
Bundy JL, Judson R, Williams AJ, Grulke C, Shah I, Everett LJ. Predicting molecular initiating events using chemical target annotations and gene expression. BioData Min 2022; 15:7. [PMID: 35246223 PMCID: PMC8895536 DOI: 10.1186/s13040-022-00292-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 02/10/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The advent of high-throughput transcriptomic screening technologies has resulted in a wealth of publicly available gene expression data associated with chemical treatments. From a regulatory perspective, data sets that cover a large chemical space and contain reference chemicals offer utility for the prediction of molecular initiating events associated with chemical exposure. Here, we integrate data from a large compendium of transcriptomic responses to chemical exposure with a comprehensive database of chemical-protein associations to train binary classifiers that predict mechanism(s) of action from transcriptomic responses. First, we linked reference chemicals present in the LINCS L1000 gene expression data collection to chemical identifiers in RefChemDB, a database of chemical-protein interactions. Next, we trained binary classifiers on MCF7 human breast cancer cell line derived gene expression profiles and chemical-protein labels using six classification algorithms to identify optimal analysis parameters. To validate classifier accuracy, we used holdout data sets, training-excluded reference chemicals, and empirical significance testing of null models derived from permuted chemical-protein associations. To identify classifiers that have variable predicting performance across training data derived from different cellular contexts, we trained a separate set of binary classifiers on the PC3 human prostate cancer cell line. RESULTS We trained classifiers using expression data associated with chemical treatments linked to 51 molecular initiating events. This analysis identified and validated 9 high-performing classifiers with empirical p-values lower than 0.05 and internal accuracies ranging from 0.73 to 0.94 and holdout accuracies of 0.68 to 0.92. High-ranking predictions for training-excluded reference chemicals demonstrating that predictive accuracy extends beyond the set of chemicals used in classifier training. To explore differences in classifier performance as a function of training data cellular context, MCF7-trained classifier accuracies were compared to classifiers trained on the PC3 gene expression data for the same molecular initiating events. CONCLUSIONS This methodology can offer insight in prioritizing candidate perturbagens of interest for targeted screens. This approach can also help guide the selection of relevant cellular contexts for screening classes of candidate perturbagens using cell line specific model performance.
Collapse
Affiliation(s)
- Joseph L Bundy
- Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, 109 T.W. Alexander Drive, Durham, NC, 27709, USA
| | - Richard Judson
- Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, 109 T.W. Alexander Drive, Durham, NC, 27709, USA
| | - Antony J Williams
- Chemical Characterization and Exposure Division, Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, 109 T.W. Alexander Drive, Durham, NC, 27709, USA
| | - Chris Grulke
- Chemical Characterization and Exposure Division, Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, 109 T.W. Alexander Drive, Durham, NC, 27709, USA
| | - Imran Shah
- Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, 109 T.W. Alexander Drive, Durham, NC, 27709, USA
| | - Logan J Everett
- Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, 109 T.W. Alexander Drive, Durham, NC, 27709, USA.
| |
Collapse
|
9
|
Trapotsi MA, Hosseini-Gerami L, Bender A. Computational analyses of mechanism of action (MoA): data, methods and integration. RSC Chem Biol 2022; 3:170-200. [PMID: 35360890 PMCID: PMC8827085 DOI: 10.1039/d1cb00069a] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 12/09/2021] [Indexed: 12/15/2022] Open
Abstract
The elucidation of a compound's Mechanism of Action (MoA) is a challenging task in the drug discovery process, but it is important in order to rationalise phenotypic findings and to anticipate potential side-effects. Bioinformatic approaches, advances in machine learning techniques and the increasing deposition of high-throughput data in public databases have significantly contributed to recent advances in the field, but it is not straightforward to decide which data and methods are most suitable to use in a given case. In this review, we focus on these methods and data and their applications in generating MoA hypotheses for subsequent experimental validation. We discuss compound-specific data such as -omics, cell morphology and bioactivity data, as well as commonly used supplementary prior knowledge such as network and pathway data, and provide information on databases where this data can be accessed. In terms of methodologies, we discuss both well-established methods (connectivity mapping, pathway enrichment) as well as more developing methods (neural networks and multi-omics integration). Finally, we review case studies where the MoA of a compound was successfully suggested from computational analysis by incorporating multiple data modalities and/or methodologies. Our aim for this review is to provide researchers with insights into the benefits and drawbacks of both the data and methods in terms of level of understanding, biases and interpretation - and to highlight future avenues of investigation which we foresee will improve the field of MoA elucidation, including greater public access to -omics data and methodologies which are capable of data integration.
Collapse
Affiliation(s)
- Maria-Anna Trapotsi
- Centre for Molecular Informatics, Yusuf Hamied Department of Chemistry, University of Cambridge UK
| | - Layla Hosseini-Gerami
- Centre for Molecular Informatics, Yusuf Hamied Department of Chemistry, University of Cambridge UK
| | - Andreas Bender
- Centre for Molecular Informatics, Yusuf Hamied Department of Chemistry, University of Cambridge UK
| |
Collapse
|
10
|
Janowska-Sejda EI, Adeleye Y, Currie RA. Exploration of the DARTable Genome- a Resource Enabling Data-Driven NAMs for Developmental and Reproductive Toxicity Prediction. FRONTIERS IN TOXICOLOGY 2022; 3:806311. [PMID: 35295108 PMCID: PMC8915813 DOI: 10.3389/ftox.2021.806311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/22/2021] [Indexed: 12/12/2022] Open
Abstract
The identification of developmental and reproductive toxicity (DART) is a critical component of toxicological evaluations of chemical safety. Adverse Outcome Pathways (AOPs) provide a framework to describe biological processes leading to a toxic effect and can provide insights in understanding the mechanisms underlying toxicological endpoints and aid the development of new approach methods (NAMs). Integrated approaches to testing and assessment (IATA) can be developed based on AOP knowledge and can serve as pragmatic approaches to chemical hazard characterization using NAMs. However, DART effects remain difficult to predict given the diversity of biological mechanisms operating during ontogenesis and consequently, the considerable number of potential molecular initiating events (MIEs) that might trigger a DART Adverse Outcome (DART AO). Consequently, two challenges that need to be overcome to create an AOP-based DART IATA are having sufficient knowledge of relevant biology and using this knowledge to determine the appropriate selection of cell systems that provide sufficient coverage of that biology. The wealth of modern biological and bioinformatics data can be used to provide this knowledge. Here we demonstrate the utility of bioinformatics analyses to address these questions. We integrated known DART MIEs with gene-developmental phenotype information to curate the hypothetical human DARTable genome (HDG, ∼5 k genes) which represents the comprehensive set of biomarkers for DART. Using network analysis of the human interactome, we show that HDG genes have distinct connectivity compared to other genes. HDG genes have higher node degree with lower neighborhood connectivity, betweenness centralities and average shortest path length. Therefore, HDG is highly connected to itself and to the wider network and not only to their local community. Also, by comparison with the Druggable Genome we show how the HDG can be prioritized to identify potential MIEs based on potential to interact with small molecules. We demonstrate how the HDG in combination with gene expression data can be used to select a panel of relevant cell lines (RD-1, OVCAR-3) for inclusion in an IATA and conclude that bioinformatic analyses can provide the necessary insights and serve as a resource for the development of a screening panel for a DART IATA.
Collapse
|
11
|
Timmons JA, Anighoro A, Brogan RJ, Stahl J, Wahlestedt C, Farquhar DG, Taylor-King J, Volmar CH, Kraus WE, Phillips SM. A human-based multi-gene signature enables quantitative drug repurposing for metabolic disease. eLife 2022; 11:68832. [PMID: 35037854 PMCID: PMC8763401 DOI: 10.7554/elife.68832] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 11/26/2021] [Indexed: 12/22/2022] Open
Abstract
Insulin resistance (IR) contributes to the pathophysiology of diabetes, dementia, viral infection, and cardiovascular disease. Drug repurposing (DR) may identify treatments for IR; however, barriers include uncertainty whether in vitro transcriptomic assays yield quantitative pharmacological data, or how to optimise assay design to best reflect in vivo human disease. We developed a clinical-based human tissue IR signature by combining lifestyle-mediated treatment responses (>500 human adipose and muscle biopsies) with biomarkers of disease status (fasting IR from >1200 biopsies). The assay identified a chemically diverse set of >130 positively acting compounds, highly enriched in true positives, that targeted 73 proteins regulating IR pathways. Our multi-gene RNA assay score reflected the quantitative pharmacological properties of a set of epidermal growth factor receptor-related tyrosine kinase inhibitors, providing insight into drug target specificity; an observation supported by deep learning-based genome-wide predicted pharmacology. Several drugs identified are suitable for evaluation in patients, particularly those with either acute or severe chronic IR.
Collapse
Affiliation(s)
- James A Timmons
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.,Augur Precision Medicine LTD, Stirling, United Kingdom
| | | | | | - Jack Stahl
- Center for Therapeutic Innovation, Miller School of Medicine, University of Miami, Miami, United States
| | - Claes Wahlestedt
- Center for Therapeutic Innovation, Miller School of Medicine, University of Miami, Miami, United States
| | | | | | - Claude-Henry Volmar
- Center for Therapeutic Innovation, Miller School of Medicine, University of Miami, Miami, United States
| | | | - Stuart M Phillips
- Faculty of Science, Kinesiology, McMaster University, Hamilton, Canada
| |
Collapse
|
12
|
Recent advances in drug repurposing using machine learning. Curr Opin Chem Biol 2021; 65:74-84. [PMID: 34274565 DOI: 10.1016/j.cbpa.2021.06.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 12/11/2022]
Abstract
Drug repurposing aims to find new uses for already existing and approved drugs. We now provide a brief overview of recent developments in drug repurposing using machine learning alongside other computational approaches for comparison. We also highlight several applications for cancer using kinase inhibitors, Alzheimer's disease as well as COVID-19.
Collapse
|
13
|
Trapotsi MA, Mervin LH, Afzal AM, Sturm N, Engkvist O, Barrett IP, Bender A. Comparison of Chemical Structure and Cell Morphology Information for Multitask Bioactivity Predictions. J Chem Inf Model 2021; 61:1444-1456. [PMID: 33661004 DOI: 10.1021/acs.jcim.0c00864] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The understanding of the mechanism-of-action (MoA) of compounds and the prediction of potential drug targets play an important role in small-molecule drug discovery. The aim of this work was to compare chemical and cell morphology information for bioactivity prediction. The comparison was performed using bioactivity data from the ExCAPE database, image data (in the form of CellProfiler features) from the Cell Painting data set (the largest publicly available data set of cell images with ∼30,000 compound perturbations), and extended connectivity fingerprints (ECFPs) using the multitask Bayesian matrix factorization (BMF) approach Macau. We found that the BMF Macau and random forest (RF) performance were overall similar when ECFPs were used as compound descriptors. However, BMF Macau outperformed RF in 159 out of 224 targets (71%) when image data were used as compound information. Using BMF Macau, 100 (corresponding to about 45%) and 90 (about 40%) of the 224 targets were predicted with high predictive performance (AUC > 0.8) with ECFP data and image data as side information, respectively. There were targets better predicted by image data as side information, such as β-catenin, and others better predicted by fingerprint-based side information, such as proteins belonging to the G-protein-Coupled Receptor 1 family, which could be rationalized from the underlying data distributions in each descriptor domain. In conclusion, both cell morphology changes and chemical structure information contain information about compound bioactivity, which is also partially complementary, and can hence contribute to in silico MoA analysis.
Collapse
Affiliation(s)
- Maria-Anna Trapotsi
- Department of Chemistry, Centre for Molecular Informatics, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| | - Lewis H Mervin
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 0WG, U.K
| | - Avid M Afzal
- Data Sciences & Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 0WG, U.K
| | - Noé Sturm
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Gothenburg SE-43183, Sweden
| | - Ola Engkvist
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Gothenburg SE-43183, Sweden
| | - Ian P Barrett
- Data Sciences & Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 0WG, U.K
| | - Andreas Bender
- Department of Chemistry, Centre for Molecular Informatics, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| |
Collapse
|
14
|
Adeowo FY, Lawal MM, Kumalo HM. Design and Development of Cholinesterase Dual Inhibitors towards Alzheimer's Disease Treatment: A Focus on Recent Contributions from Computational and Theoretical Perspective. ChemistrySelect 2020. [DOI: 10.1002/slct.202003573] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Fatima Y. Adeowo
- Discipline of Medical Biochemistry School of Laboratory Medicine and Medical Science University of KwaZulu-Natal Durban 4001 South Africa
| | - Monsurat M. Lawal
- Discipline of Medical Biochemistry School of Laboratory Medicine and Medical Science University of KwaZulu-Natal Durban 4001 South Africa
| | - Hezekiel M. Kumalo
- Discipline of Medical Biochemistry School of Laboratory Medicine and Medical Science University of KwaZulu-Natal Durban 4001 South Africa
| |
Collapse
|