1
|
Seal S, Mahale M, García-Ortegón M, Joshi CK, Hosseini-Gerami L, Beatson A, Greenig M, Shekhar M, Patra A, Weis C, Mehrjou A, Badré A, Paisley B, Lowe R, Singh S, Shah F, Johannesson B, Williams D, Rouquie D, Clevert DA, Schwab P, Richmond N, Nicolaou CA, Gonzalez RJ, Naven R, Schramm C, Vidler LR, Mansouri K, Walters WP, Wilk DD, Spjuth O, Carpenter AE, Bender A. Machine Learning for Toxicity Prediction Using Chemical Structures: Pillars for Success in the Real World. Chem Res Toxicol 2025. [PMID: 40314361 DOI: 10.1021/acs.chemrestox.5c00033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Machine learning (ML) is increasingly valuable for predicting molecular properties and toxicity in drug discovery. However, toxicity-related end points have always been challenging to evaluate experimentally with respect to in vivo translation due to the required resources for human and animal studies; this has impacted data availability in the field. ML can augment or even potentially replace traditional experimental processes depending on the project phase and specific goals of the prediction. For instance, models can be used to select promising compounds for on-target effects or to deselect those with undesirable characteristics (e.g., off-target or ineffective due to unfavorable pharmacokinetics). However, reliance on ML is not without risks, due to biases stemming from nonrepresentative training data, incompatible choice of algorithm to represent the underlying data, or poor model building and validation approaches. This might lead to inaccurate predictions, misinterpretation of the confidence in ML predictions, and ultimately suboptimal decision-making. Hence, understanding the predictive validity of ML models is of utmost importance to enable faster drug development timelines while improving the quality of decisions. This perspective emphasizes the need to enhance the understanding and application of machine learning models in drug discovery, focusing on well-defined data sets for toxicity prediction based on small molecule structures. We focus on five crucial pillars for success with ML-driven molecular property and toxicity prediction: (1) data set selection, (2) structural representations, (3) model algorithm, (4) model validation, and (5) translation of predictions to decision-making. Understanding these key pillars will foster collaboration and coordination between ML researchers and toxicologists, which will help to advance drug discovery and development.
Collapse
Affiliation(s)
- Srijit Seal
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
| | - Manas Mahale
- Department of Pharmaceutical Chemistry, Bombay College of Pharmacy, Mumbai 400098, India
| | | | - Chaitanya K Joshi
- Department of Computer Science and Technology, University of Cambridge, Cambridge CB3 0FD, U.K
| | | | - Alex Beatson
- Axiom Bio, San Francisco, California 94107, United States
| | - Matthew Greenig
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
| | - Mrinal Shekhar
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | | | | | | | - Adrien Badré
- Novartis Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Brianna Paisley
- Eli Lilly & Company, Indianapolis, Indiana 46285, United States
| | | | - Shantanu Singh
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Falgun Shah
- Non Clinical Drug Safety, Merck Inc., West Point, Pennsylvania 19486, United States
| | | | | | - David Rouquie
- Toxicology Data Science, Bayer SAS Crop Science Division, Valbonne Sophia-Antipolis 06560, France
| | - Djork-Arné Clevert
- Pfizer, Worldwide Research, Development and Medical, Machine Learning & Computational Sciences, Berlin 10922, Germany
| | | | | | - Christos A Nicolaou
- Computational Drug Design, Digital Science & Innovation, Novo Nordisk US R&D, Lexington, Massachusetts 02421, United States
| | - Raymond J Gonzalez
- Non Clinical Drug Safety, Merck Inc., West Point, Pennsylvania 19486, United States
| | - Russell Naven
- Novartis Biomedical Research, Cambridge, Massachusetts 02139, United States
| | | | | | - Kamel Mansouri
- NIH/NIEHS/DTT/NICEATM, Research Triangle Park, North Carolina 27709, United States
| | | | | | - Ola Spjuth
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Uppsala 751 24, Sweden
- Phenaros Pharmaceuticals AB, Uppsala 75239, Sweden
| | - Anne E Carpenter
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Andreas Bender
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
- College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| |
Collapse
|
2
|
Alam F, Mohammed Alnazzawi TS, Mehmood R, Al-maghthawi A. A Review of the Applications, Benefits, and Challenges of Generative AI for Sustainable Toxicology. Curr Res Toxicol 2025; 8:100232. [PMID: 40331045 PMCID: PMC12051651 DOI: 10.1016/j.crtox.2025.100232] [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: 10/20/2024] [Revised: 03/10/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Sustainable toxicology is vital for living species and the environment because it guarantees the safety, efficacy, and regulatory compliance of drugs, treatments, vaccines, and chemicals in living organisms and the environment. Conventional toxicological methods often lack sustainability as they are costly, time-consuming, and sometimes inaccurate. It means delays in producing new drugs, vaccines, and treatments and understanding the adverse effects of the chemicals on the environment. To address these challenges, the healthcare sector must leverage the power of the Generative-AI (GenAI) paradigm. This paper aims to help understand how the healthcare field can be revolutionized in multiple ways by using GenAI to facilitate sustainable toxicological developments. This paper first reviews the present literature and identifies the possible classes of GenAI that can be applied to toxicology. A generalized and holistic visualization of various toxicological processes powered by GenAI is presented in tandem. The paper discussed toxicological risk assessment and management, spotlighting how global agencies and organizations are forming policies to standardize and regulate AI-related development, such as GenAI, in these fields. The paper identifies and discusses the advantages and challenges of GenAI in toxicology. Further, the paper outlines how GenAI empowers Conversational-AI, which will be critical for highly tailored toxicological solutions. This review will help to develop a comprehensive understanding of the impacts and future potential of GenAI in the field of toxicology. The knowledge gained can be applied to create sustainable GenAI applications for various problems in toxicology, ultimately benefiting our societies and the environment.
Collapse
Affiliation(s)
- Furqan Alam
- Faculty of Computing and Information Technology (FoCIT), Sohar University, Sohar 311, Oman
| | - Tahani Saleh Mohammed Alnazzawi
- Department of Computer Science, College of Computer Science and Engineering, Taibah University, Madinah 41477, Kingdom of Saudi Arabia
| | - Rashid Mehmood
- Faculty of Computer Science and Information Systems, Islamic University Madinah, Madinah 42351, Kingdom of Saudi Arabia
| | - Ahmed Al-maghthawi
- Department of Computer Science, College of Science & Art at Mahayil, King Khalid University, Abha 62529, Kingdom of Saudi Arabia
| |
Collapse
|
3
|
Gangwal A, Lavecchia A. Artificial intelligence in preclinical research: enhancing digital twins and organ-on-chip to reduce animal testing. Drug Discov Today 2025; 30:104360. [PMID: 40252989 DOI: 10.1016/j.drudis.2025.104360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/28/2025] [Accepted: 04/10/2025] [Indexed: 04/21/2025]
Abstract
Artificial intelligence (AI) is reshaping preclinical drug research offering innovative alternatives to traditional animal testing. Advanced techniques, including machine learning (ML), deep learning (DL), AI-powered digital twins (DTs), and AI-enhanced organ-on-a-chip (OoC) platforms, enable precise simulations of complex biological systems. AI plays a critical role in overcoming the limitations of DTs and OoC, improving their predictive power and scalability. These technologies facilitate early-stage, reliable evaluations of drug safety and efficacy, addressing ethical concerns, reducing costs, and accelerating drug development while adhering to the 3Rs principle (Replace, Reduce, Refine). By integrating AI with these advanced models, preclinical research can achieve greater accuracy and efficiency in drug discovery. This review examines the transformative impact of AI in preclinical research, highlighting its advancements, challenges, and the critical steps needed to establish AI as a cornerstone of ethical and efficient drug discovery.
Collapse
Affiliation(s)
- Amit Gangwal
- Department of Natural Product Chemistry, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule 424001 Maharashtra, India
| | - Antonio Lavecchia
- "Drug Discovery" Laboratory, Department of Pharmacy, University of Naples Federico II, I-80131 Naples, Italy.
| |
Collapse
|
4
|
Tropsha A, Martin HJ, Cherkasov A. The Six Ds of Exponentials and drug discovery: A path toward reversing Eroom's law. Drug Discov Today 2025; 30:104341. [PMID: 40122449 PMCID: PMC12043357 DOI: 10.1016/j.drudis.2025.104341] [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: 01/21/2025] [Revised: 03/09/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Many technological sectors underwent recent exponential growth because of digital disruption, a phenomenon Peter Diamantis characterized as the 'Six Ds of Exponentials': digitization, deception, disruption, demonetization, dematerialization, and democratization. In contrast, drug discovery has been marked by rising costs and modest growth, if any, of annual drug approvals. We argue that the exponential growth of drug discovery can be also achieved through digital disruption brought by data expansion, mature artificial intelligence (AI), automation of experiments, public-private partnerships, and open science. We detected the emergence of all 'Six Ds of Exponentials' within modern drug discovery and discuss how each of the 'Six Ds' can further empower the field and forcefully address the societal demand for novel, potent, affordable, and accessible medicines.
Collapse
Affiliation(s)
- Alexander Tropsha
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Holli-Joi Martin
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Artem Cherkasov
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
5
|
Huang L, Duan Q, Liu Y, Wu Y, Li Z, Guo Z, Liu M, Lu X, Wang P, Liu F, Ren F, Li C, Wang J, Huang Y, Yan B, Kioumourtzoglou MA, Kinney PL. Artificial intelligence: A key fulcrum for addressing complex environmental health issues. ENVIRONMENT INTERNATIONAL 2025; 198:109389. [PMID: 40121790 DOI: 10.1016/j.envint.2025.109389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/16/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Environmental health (EH) is a complex and interdisciplinary field dedicated to the examination of environmental behaviours, toxicological effects, health risks, and strategies for mitigating harmful environmental factors. Traditional EH research investigates correlations between risk factors and health outcomes through control variables, but this route is difficult to address complex EH issue. Artificial intelligence (AI) technology not only has accelerated the innovation of the scientific research paradigm but also has become an important tool for solving complex EH problems. However, the in-depth and comprehensive implementation of AI in the field of EH still faces many barriers, such as model generalizability, data privacy protection, algorithm transparency, and regulatory and ethical issues. This review focuses on the compound exposures of EH and explores the potential, challenges, and development directions of AI in four key phases of EH research: (1) data collection, fusion, and management, (2) hazard identification and screening, (3) risk modeling and assessment and (4) EH management. It is not difficult to see that in the future, artificial intelligence technology will inevitably carry out multidimensional simulation of complex exposure factors through multi-mode data fusion, so as to achieve accurate identification of environmental health risks, and eventually become an efficient tool for global environmental health management. This review will help researchers re-examine this strategy and provide a reference for AI to solve complex exposure problems.
Collapse
Affiliation(s)
- Lei Huang
- State Key Laboratory of Water Pollution Control and Green Resource Recycling, School of the Environment, Nanjing University, Nanjing 210023, China; Basic Science Center for Energy and Climate Change, Beijing 100081, China.
| | - Qiannan Duan
- Shaanxi Key Laboratory of Earth Surface System and Environmental Carrying Capacity, College of Urban and Environmental Sciences, Northwest University, Xi'an 710127, China.
| | - Yuxin Liu
- State Key Laboratory of Water Pollution Control and Green Resource Recycling, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Yangyang Wu
- State Key Laboratory of Water Pollution Control and Green Resource Recycling, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Zenghui Li
- State Key Laboratory of Water Pollution Control and Green Resource Recycling, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Zhao Guo
- State Key Laboratory of Water Pollution Control and Green Resource Recycling, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Mingliang Liu
- State Key Laboratory of Water Pollution Control and Green Resource Recycling, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Xiaowei Lu
- State Key Laboratory of Water Pollution Control and Green Resource Recycling, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Peng Wang
- Faculty of Civil Engineering and Mechanics, Jiangsu University, Zhenjiang 212013, China
| | - Fan Liu
- State Key Laboratory of Water Pollution Control and Green Resource Recycling, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Futian Ren
- State Key Laboratory of Water Pollution Control and Green Resource Recycling, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Chen Li
- State Key Laboratory of Water Pollution Control and Green Resource Recycling, School of the Environment, Nanjing University, Nanjing 210023, China; Medical School, Nanjing University, Nanjing 210093, China
| | - Jiaming Wang
- State Key Laboratory of Water Pollution Control and Green Resource Recycling, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Yujia Huang
- State Key Laboratory of Water Pollution Control and Green Resource Recycling, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Beizhan Yan
- Lamont-Doherty Earth Observatory, Columbia University, New York, USA
| | | | | |
Collapse
|
6
|
Bai C, Wu L, Li R, Cao Y, He S, Bo X. Machine Learning-Enabled Drug-Induced Toxicity Prediction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413405. [PMID: 39899688 PMCID: PMC12021114 DOI: 10.1002/advs.202413405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/25/2024] [Indexed: 02/05/2025]
Abstract
Unexpected toxicity has become a significant obstacle to drug candidate development, accounting for 30% of drug discovery failures. Traditional toxicity assessment through animal testing is costly and time-consuming. Big data and artificial intelligence (AI), especially machine learning (ML), are robustly contributing to innovation and progress in toxicology research. However, the optimal AI model for different types of toxicity usually varies, making it essential to conduct comparative analyses of AI methods across toxicity domains. The diverse data sources also pose challenges for researchers focusing on specific toxicity studies. In this review, 10 categories of drug-induced toxicity is examined, summarizing the characteristics and applicable ML models, including both predictive and interpretable algorithms, striking a balance between breadth and depth. Key databases and tools used in toxicity prediction are also highlighted, including toxicology, chemical, multi-omics, and benchmark databases, organized by their focus and function to clarify their roles in drug-induced toxicity prediction. Finally, strategies to turn challenges into opportunities are analyzed and discussed. This review may provide researchers with a valuable reference for understanding and utilizing the available resources to bridge prediction and mechanistic insights, and further advance the application of ML in drugs-induced toxicity prediction.
Collapse
Affiliation(s)
- Changsen Bai
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- Department of Advanced & Interdisciplinary BiotechnologyAcademy of Military Medical SciencesBeijing100850China
- Tianjin Medical University Cancer Institute and HospitalTianjin300060China
| | - Lianlian Wu
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- Department of Advanced & Interdisciplinary BiotechnologyAcademy of Military Medical SciencesBeijing100850China
| | - Ruijiang Li
- Department of Advanced & Interdisciplinary BiotechnologyAcademy of Military Medical SciencesBeijing100850China
| | - Yang Cao
- Department of Environmental MedicineAcademy of Military Medical SciencesTianjin300050China
| | - Song He
- Department of Advanced & Interdisciplinary BiotechnologyAcademy of Military Medical SciencesBeijing100850China
| | - Xiaochen Bo
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- Department of Advanced & Interdisciplinary BiotechnologyAcademy of Military Medical SciencesBeijing100850China
| |
Collapse
|
7
|
Meier MJ, Harrill J, Johnson K, Thomas RS, Tong W, Rager JE, Yauk CL. Progress in toxicogenomics to protect human health. Nat Rev Genet 2025; 26:105-122. [PMID: 39223311 DOI: 10.1038/s41576-024-00767-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
Toxicogenomics measures molecular features, such as transcripts, proteins, metabolites and epigenomic modifications, to understand and predict the toxicological effects of environmental and pharmaceutical exposures. Transcriptomics has become an integral tool in contemporary toxicology research owing to innovations in gene expression profiling that can provide mechanistic and quantitative information at scale. These data can be used to predict toxicological hazards through the use of transcriptomic biomarkers, network inference analyses, pattern-matching approaches and artificial intelligence. Furthermore, emerging approaches, such as high-throughput dose-response modelling, can leverage toxicogenomic data for human health protection even in the absence of predicting specific hazards. Finally, single-cell transcriptomics and multi-omics provide detailed insights into toxicological mechanisms. Here, we review the progress since the inception of toxicogenomics in applying transcriptomics towards toxicology testing and highlight advances that are transforming risk assessment.
Collapse
Affiliation(s)
- Matthew J Meier
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Joshua Harrill
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC, USA
| | - Kamin Johnson
- Predictive Safety Center, Corteva Agriscience, Indianapolis, IN, USA
| | - Russell S Thomas
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC, USA
| | - Weida Tong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, United States Food and Drug Administration, Jefferson, AR, USA
- Curriculum in Toxicology & Environmental Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Julia E Rager
- Curriculum in Toxicology & Environmental Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- The Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, The University of North Carolina, Chapel Hill, NC, USA
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Carole L Yauk
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
8
|
Guimarães AI. Are Animal Models Necessary? Exploring (Dis)advantages and Alternatives. Eur J Neurosci 2025; 61:e16651. [PMID: 39780289 DOI: 10.1111/ejn.16651] [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: 10/17/2024] [Revised: 12/04/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025]
Abstract
Animal models have been crucial for scientific development, allowing researchers to understand the underlying mechanisms of various human conditions, and are far from becoming obsolete in scientific research. However, the ethics of animal experimentation has been a prevalent question between both experts and nonexperts. This essay tackles the advantages and disadvantages of the usage of animal models while delving into new alternatives that have emerged in light of contemporary science.
Collapse
Affiliation(s)
- Ana Isabel Guimarães
- Pharmacology and Neurobiology Laboratory of the Immunophysiology and Pharmacology Department, School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
- Center for Drug Discovery and Innovative Medicines (MedInUP), School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
| |
Collapse
|
9
|
Rao M, Nassiri V, Srivastava S, Yang A, Brar S, McDuffie E, Sachs C. Artificial Intelligence and Machine Learning Models for Predicting Drug-Induced Kidney Injury in Small Molecules. Pharmaceuticals (Basel) 2024; 17:1550. [PMID: 39598459 PMCID: PMC11597314 DOI: 10.3390/ph17111550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/09/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND/OBJECTIVES Drug-Induced Kidney Injury (DIKI) presents a significant challenge in drug development, often leading to clinical-stage failures. The early prediction of DIKI risk can improve drug safety and development efficiency. Existing models tend to focus on physicochemical properties alone, often overlooking drug-target interactions crucial for DIKI. This study introduces an AI/ML (artificial intelligence/machine learning) model that integrates both physicochemical properties and off-target interactions to enhance DIKI prediction. METHODS We compiled a dataset of 360 FDA-classified compounds (231 non-nephrotoxic and 129 nephrotoxic) and predicted 6064 off-target interactions, 59% of which were validated in vitro. We also calculated 55 physicochemical properties for these compounds. Machine learning (ML) models were developed using four algorithms: Ridge Logistic Regression (RLR), Support Vector Machine (SVM), Random Forest (RF), and Neural Network (NN). These models were then combined into an ensemble model for enhanced performance. RESULTS The ensemble model achieved an ROC-AUC of 0.86, with a sensitivity and specificity of 0.79 and 0.78, respectively. The key predictive features included 38 off-target interactions and physicochemical properties such as the number of metabolites, polar surface area (PSA), pKa, and fraction of Sp3-hybridized carbons (fsp3). These features effectively distinguished DIKI from non-DIKI compounds. CONCLUSIONS The integrated model, which combines both physicochemical properties and off-target interaction data, significantly improved DIKI prediction accuracy compared to models that rely on either data type alone. This AI/ML model provides a promising early screening tool for identifying compounds with lower DIKI risk, facilitating safer drug development.
Collapse
Affiliation(s)
- Mohan Rao
- Preclinical and Clinical Pharmacology and Chemistry, Neurocrine Biosciences, San Diego, CA 92130, USA (C.S.)
| | - Vahid Nassiri
- Open Analytics NV, Jupiterstraat 20, 2600 Antwerp, Belgium;
| | - Sanjay Srivastava
- Preclinical and Clinical Pharmacology and Chemistry, Neurocrine Biosciences, San Diego, CA 92130, USA (C.S.)
| | - Amy Yang
- Preclinical and Clinical Pharmacology and Chemistry, Neurocrine Biosciences, San Diego, CA 92130, USA (C.S.)
| | - Satjit Brar
- Preclinical and Clinical Pharmacology and Chemistry, Neurocrine Biosciences, San Diego, CA 92130, USA (C.S.)
| | - Eric McDuffie
- Preclinical and Clinical Pharmacology and Chemistry, Neurocrine Biosciences, San Diego, CA 92130, USA (C.S.)
| | - Clifford Sachs
- Preclinical and Clinical Pharmacology and Chemistry, Neurocrine Biosciences, San Diego, CA 92130, USA (C.S.)
| |
Collapse
|
10
|
Li T, Chen X, Tong W. Bridging organ transcriptomics for advancing multiple organ toxicity assessment with a generative AI approach. NPJ Digit Med 2024; 7:310. [PMID: 39501092 PMCID: PMC11538515 DOI: 10.1038/s41746-024-01317-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 10/25/2024] [Indexed: 11/08/2024] Open
Abstract
Translational research in toxicology has significantly benefited from transcriptomic profiling, particularly in drug safety. However, its application has predominantly focused on limited organs, notably the liver, due to resource constraints. This paper presents TransTox, an innovative AI model using a generative adversarial network (GAN) method to facilitate the bidirectional translation of transcriptomic profiles between the liver and kidney under drug treatment. TransTox demonstrates robust performance, validated across independent datasets and laboratories. First, the concordance between real experimental data and synthetic data generated by TransTox was demonstrated in characterizing toxicity mechanisms compared to real experimental settings. Second, TransTox proved valuable in gene expression predictive models, where synthetic data could be used to develop gene expression predictive models or serve as "digital twins" for diagnostic applications. The TransTox approach holds the potential for multi-organ toxicity assessment with AI and advancing the field of precision toxicology.
Collapse
Affiliation(s)
- Ting Li
- FDA National Center for Toxicological Research, Jefferson, AR, USA
| | - Xi Chen
- FDA National Center for Toxicological Research, Jefferson, AR, USA
| | - Weida Tong
- FDA National Center for Toxicological Research, Jefferson, AR, USA.
| |
Collapse
|
11
|
Langan LM, Paparella M, Burden N, Constantine L, Margiotta-Casaluci L, Miller TH, Moe SJ, Owen SF, Schaffert A, Sikanen T. Big Question to Developing Solutions: A Decade of Progress in the Development of Aquatic New Approach Methodologies from 2012 to 2022. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2024; 43:559-574. [PMID: 36722131 PMCID: PMC10390655 DOI: 10.1002/etc.5578] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/26/2022] [Accepted: 01/24/2023] [Indexed: 06/18/2023]
Abstract
In 2012, 20 key questions related to hazard and exposure assessment and environmental and health risks of pharmaceuticals and personal care products in the natural environment were identified. A decade later, this article examines the current level of knowledge around one of the lowest-ranking questions at that time, number 19: "Can nonanimal testing methods be developed that will provide equivalent or better hazard data compared with current in vivo methods?" The inclusion of alternative methods that replace, reduce, or refine animal testing within the regulatory context of risk and hazard assessment of chemicals generally faces many hurdles, although this varies both by organism (human-centric vs. other), sector, and geographical region or country. Focusing on the past 10 years, only works that might reasonably be considered to contribute to advancements in the field of aquatic environmental risk assessment are highlighted. Particular attention is paid to methods of contemporary interest and importance, representing progress in (1) the development of methods which provide equivalent or better data compared with current in vivo methods such as bioaccumulation, (2) weight of evidence, or (3) -omic-based applications. Evolution and convergence of these risk assessment areas offer the basis for fundamental frameshifts in how data are collated and used for the protection of taxa across the breadth of the aquatic environment. Looking to the future, we are at a tipping point, with a need for a global and inclusive approach to establish consensus. Bringing together these methods (both new and old) for regulatory assessment and decision-making will require a concerted effort and orchestration. Environ Toxicol Chem 2024;43:559-574. © 2023 The Authors. Environmental Toxicology and Chemistry published by Wiley Periodicals LLC on behalf of SETAC.
Collapse
Affiliation(s)
- Laura M Langan
- Department of Environmental Science, Baylor University, One Bear Place #97266, Waco, TX, 76798, USA
| | - Martin Paparella
- Department of Medical Biochemistry, Medical University of Innsbruck, Innrain 80, 6020 Innsbruck, Austria
| | - Natalie Burden
- National Centre for the 3Rs (NC3Rs), Gibbs Building, 215 Euston Road, London NW1 2BE, UK
| | | | - Luigi Margiotta-Casaluci
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer and Pharmaceutical Sciences, King’s College London, London SE1 9NQ, UK
| | - Thomas H. Miller
- Centre for Pollution Research & Policy, Environmental Sciences, Brunel University London, London, UK
| | - S. Jannicke Moe
- Norwegian Institute for Water Research, Økernveien 94, 0579 Oslo, Norway
| | - Stewart F. Owen
- AstraZeneca, Global Sustainability, Macclesfield, Cheshire SK10 2NA, UK
| | - Alexandra Schaffert
- Department of Medical Biochemistry, Medical University of Innsbruck, Innrain 80, 6020 Innsbruck, Austria
| | - Tiina Sikanen
- Faculty of Pharmacy and Helsinki Institute of Sustainability Science, University of Helsinki, Yliopistonkatu 3, Helsinki, 00100, Finland
| |
Collapse
|
12
|
Kaplan BLF, Hoberman AM, Slikker W, Smith MA, Corsini E, Knudsen TB, Marty MS, Sobrian SK, Fitzpatrick SC, Ratner MH, Mendrick DL. Protecting Human and Animal Health: The Road from Animal Models to New Approach Methods. Pharmacol Rev 2024; 76:251-266. [PMID: 38351072 PMCID: PMC10877708 DOI: 10.1124/pharmrev.123.000967] [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: 06/26/2023] [Revised: 10/18/2023] [Accepted: 12/01/2023] [Indexed: 02/16/2024] Open
Abstract
Animals and animal models have been invaluable for our current understanding of human and animal biology, including physiology, pharmacology, biochemistry, and disease pathology. However, there are increasing concerns with continued use of animals in basic biomedical, pharmacological, and regulatory research to provide safety assessments for drugs and chemicals. There are concerns that animals do not provide sufficient information on toxicity and/or efficacy to protect the target population, so scientists are utilizing the principles of replacement, reduction, and refinement (the 3Rs) and increasing the development and application of new approach methods (NAMs). NAMs are any technology, methodology, approach, or assay used to understand the effects and mechanisms of drugs or chemicals, with specific focus on applying the 3Rs. Although progress has been made in several areas with NAMs, complete replacement of animal models with NAMs is not yet attainable. The road to NAMs requires additional development, increased use, and, for regulatory decision making, usually formal validation. Moreover, it is likely that replacement of animal models with NAMs will require multiple assays to ensure sufficient biologic coverage. The purpose of this manuscript is to provide a balanced view of the current state of the use of animal models and NAMs as approaches to development, safety, efficacy, and toxicity testing of drugs and chemicals. Animals do not provide all needed information nor do NAMs, but each can elucidate key pieces of the puzzle of human and animal biology and contribute to the goal of protecting human and animal health. SIGNIFICANCE STATEMENT: Data from traditional animal studies have predominantly been used to inform human health safety and efficacy. Although it is unlikely that all animal studies will be able to be replaced, with the continued advancement in new approach methods (NAMs), it is possible that sometime in the future, NAMs will likely be an important component by which the discovery, efficacy, and toxicity testing of drugs and chemicals is conducted and regulatory decisions are made.
Collapse
Affiliation(s)
- Barbara L F Kaplan
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Alan M Hoberman
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - William Slikker
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Mary Alice Smith
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Emanuela Corsini
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Thomas B Knudsen
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - M Sue Marty
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Sonya K Sobrian
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Suzanne C Fitzpatrick
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Marcia H Ratner
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| | - Donna L Mendrick
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi (B.L.F.K.); Charles River Laboratories, Inc., Horsham, Pennsylvania (A.M.H.); Retired, National Center for Toxicological Research, Jefferson, Arkansas (W.S.); University of Georgia, Athens, Georgia (M.A.S.); Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti' Università degli Studi di Milano, Milan, Italy (E.C.); US Environmental Protection Agency, Research Triangle Park, North Carolina (T.B.K.); Dow, Inc., Midland, Michigan (M.S.M.); Howard University College of Medicine, Washington DC (S.K.S.); Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, Maryland (S.C.F.); Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (M.H.R.); and National Center for Toxicological Research, US Food and Drug Administration, Silver Spring, Maryland (D.L.M.)
| |
Collapse
|
13
|
Kimura H, Nakamura H, Goto T, Uchida W, Uozumi T, Nishizawa D, Shinha K, Sakagami J, Doi K. Standalone cell culture microfluidic device-based microphysiological system for automated cell observation and application in nephrotoxicity tests. LAB ON A CHIP 2024; 24:408-421. [PMID: 38131210 DOI: 10.1039/d3lc00934c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Microphysiological systems (MPS) offer an alternative method for culturing cells on microfluidic platforms to model organ functions in pharmaceutical and medical sciences. Although MPS hardware has been proposed to maintain physiological organ function through perfusion culture, no existing MPS can automatically assess cell morphology and conditions online to observe cellular dynamics in detail. Thus, with this study, we aimed to establish a practical strategy for automating cell observation and improving cell evaluation functions with low temporal resolution and throughput in MPS experiments. We developed a versatile standalone cell culture microfluidic device (SCCMD) that integrates microfluidic chips and their peripherals. This device is compliant with the ANSI/SLAS standards and has been seamlessly integrated into an existing automatic cell imaging system. This integration enables automatic cell observation with high temporal resolution in MPS experiments. Perfusion culture of human kidney proximal tubule epithelial cells using the SCCMD improves cell function. By combining the proximal tubule MPS with an existing cell imaging system, nephrotoxicity studies were successfully performed to automate morphological and material permeability evaluation. We believe that the concept of building the ANSI/SLAS-compliant-sized MPS device proposed herein and integrating it into an existing automatic cell imaging system for the online measurement of detailed cell dynamics information and improvement of throughput by automating observation operations is a novel potential research direction for MPS research.
Collapse
Affiliation(s)
- Hiroshi Kimura
- Micro/Nano Technology Center, Tokai University, Kanagawa, Japan 259-1292.
| | - Hiroko Nakamura
- Micro/Nano Technology Center, Tokai University, Kanagawa, Japan 259-1292.
| | - Tomomi Goto
- Micro/Nano Technology Center, Tokai University, Kanagawa, Japan 259-1292.
| | - Wakana Uchida
- Stem Cell Healthcare Business Unit, Nikon Corporation, Kanagawa, Japan
| | - Takayuki Uozumi
- Stem Cell Healthcare Business Unit, Nikon Corporation, Kanagawa, Japan
| | - Daniel Nishizawa
- Micro/Nano Technology Center, Tokai University, Kanagawa, Japan 259-1292.
| | - Kenta Shinha
- Micro/Nano Technology Center, Tokai University, Kanagawa, Japan 259-1292.
| | - Junko Sakagami
- Stem Cell Healthcare Business Unit, Nikon Corporation, Kanagawa, Japan
| | - Kotaro Doi
- Institute of Industrial Science, The University of Tokyo, Tokyo, Japan 153-8505
| |
Collapse
|
14
|
Mostafa F, Chen M. Computational models for predicting liver toxicity in the deep learning era. FRONTIERS IN TOXICOLOGY 2024; 5:1340860. [PMID: 38312894 PMCID: PMC10834666 DOI: 10.3389/ftox.2023.1340860] [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: 11/19/2023] [Accepted: 12/22/2023] [Indexed: 02/06/2024] Open
Abstract
Drug-induced liver injury (DILI) is a severe adverse reaction caused by drugs and may result in acute liver failure and even death. Many efforts have centered on mitigating risks associated with potential DILI in humans. Among these, quantitative structure-activity relationship (QSAR) was proven to be a valuable tool for early-stage hepatotoxicity screening. Its advantages include no requirement for physical substances and rapid delivery of results. Deep learning (DL) made rapid advancements recently and has been used for developing QSAR models. This review discusses the use of DL in predicting DILI, focusing on the development of QSAR models employing extensive chemical structure datasets alongside their corresponding DILI outcomes. We undertake a comprehensive evaluation of various DL methods, comparing with those of traditional machine learning (ML) approaches, and explore the strengths and limitations of DL techniques regarding their interpretability, scalability, and generalization. Overall, our review underscores the potential of DL methodologies to enhance DILI prediction and provides insights into future avenues for developing predictive models to mitigate DILI risk in humans.
Collapse
Affiliation(s)
- Fahad Mostafa
- Department of Mathematics and Statistics, Texas Tech University, Lubbock, TX, United States
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, United States
| | - Minjun Chen
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, United States
| |
Collapse
|
15
|
Bordukova M, Makarov N, Rodriguez-Esteban R, Schmich F, Menden MP. Generative artificial intelligence empowers digital twins in drug discovery and clinical trials. Expert Opin Drug Discov 2024; 19:33-42. [PMID: 37887266 DOI: 10.1080/17460441.2023.2273839] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 10/18/2023] [Indexed: 10/28/2023]
Abstract
INTRODUCTION The concept of Digital Twins (DTs) translated to drug development and clinical trials describes virtual representations of systems of various complexities, ranging from individual cells to entire humans, and enables in silico simulations and experiments. DTs increase the efficiency of drug discovery and development by digitalizing processes associated with high economic, ethical, or social burden. The impact is multifaceted: DT models sharpen disease understanding, support biomarker discovery and accelerate drug development, thus advancing precision medicine. One way to realize DTs is by generative artificial intelligence (AI), a cutting-edge technology that enables the creation of novel, realistic and complex data with desired properties. AREAS COVERED The authors provide a brief introduction to generative AI and describe how it facilitates the modeling of DTs. In addition, they compare existing implementations of generative AI for DTs in drug discovery and clinical trials. Finally, they discuss technical and regulatory challenges that should be addressed before DTs can transform drug discovery and clinical trials. EXPERT OPINION The current state of DTs in drug discovery and clinical trials does not exploit the entire power of generative AI yet and is limited to simulation of a small number of characteristics. Nonetheless, generative AI has the potential to transform the field by leveraging recent developments in deep learning and customizing models for the needs of scientists, physicians and patients.
Collapse
Affiliation(s)
- Maria Bordukova
- Data & Analytics, Pharmaceutical Research and Early Development, Roche Innovation Center Munich (RICM), Penzberg, Germany
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Munich, Germany
- Department of Biology, Ludwig-Maximilians University Munich, Munich, Germany
| | - Nikita Makarov
- Data & Analytics, Pharmaceutical Research and Early Development, Roche Innovation Center Munich (RICM), Penzberg, Germany
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Munich, Germany
- Department of Biology, Ludwig-Maximilians University Munich, Munich, Germany
| | - Raul Rodriguez-Esteban
- Data & Analytics, Pharmaceutical Research and Early Development, Roche Innovation Center Basel (RICB), Basel, Switzerland
| | - Fabian Schmich
- Data & Analytics, Pharmaceutical Research and Early Development, Roche Innovation Center Munich (RICM), Penzberg, Germany
| | - Michael P Menden
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Munich, Germany
- Department of Biology, Ludwig-Maximilians University Munich, Munich, Germany
- Department of Biochemistry and Pharmacology, University of Melbourne, Melbourne, Australia
- German Center for Diabetes Research (DZD e.V.), Munich, Germany
| |
Collapse
|
16
|
Jia X, Wang T, Zhu H. Advancing Computational Toxicology by Interpretable Machine Learning. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:17690-17706. [PMID: 37224004 PMCID: PMC10666545 DOI: 10.1021/acs.est.3c00653] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 05/05/2023] [Accepted: 05/05/2023] [Indexed: 05/26/2023]
Abstract
Chemical toxicity evaluations for drugs, consumer products, and environmental chemicals have a critical impact on human health. Traditional animal models to evaluate chemical toxicity are expensive, time-consuming, and often fail to detect toxicants in humans. Computational toxicology is a promising alternative approach that utilizes machine learning (ML) and deep learning (DL) techniques to predict the toxicity potentials of chemicals. Although the applications of ML- and DL-based computational models in chemical toxicity predictions are attractive, many toxicity models are "black boxes" in nature and difficult to interpret by toxicologists, which hampers the chemical risk assessments using these models. The recent progress of interpretable ML (IML) in the computer science field meets this urgent need to unveil the underlying toxicity mechanisms and elucidate the domain knowledge of toxicity models. In this review, we focused on the applications of IML in computational toxicology, including toxicity feature data, model interpretation methods, use of knowledge base frameworks in IML development, and recent applications. The challenges and future directions of IML modeling in toxicology are also discussed. We hope this review can encourage efforts in developing interpretable models with new IML algorithms that can assist new chemical assessments by illustrating toxicity mechanisms in humans.
Collapse
Affiliation(s)
- Xuelian Jia
- Department
of Chemistry and Biochemistry, Rowan University, Glassboro, New Jersey 08028, United States
| | - Tong Wang
- Department
of Chemistry and Biochemistry, Rowan University, Glassboro, New Jersey 08028, United States
| | - Hao Zhu
- Department
of Chemistry and Biochemistry, Rowan University, Glassboro, New Jersey 08028, United States
| |
Collapse
|
17
|
Chen X, Roberts R, Liu Z, Tong W. A generative adversarial network model alternative to animal studies for clinical pathology assessment. Nat Commun 2023; 14:7141. [PMID: 37932302 PMCID: PMC10628291 DOI: 10.1038/s41467-023-42933-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 10/25/2023] [Indexed: 11/08/2023] Open
Abstract
Animal studies are unavoidable in evaluating chemical and drug safety. Generative Adversarial Networks (GANs) can generate synthetic animal data by learning from the legacy animal study results, thus may serve as an alternative approach to assess untested chemicals. AnimalGAN, a GAN method to simulate 38 rat clinical pathology measures, was developed with significant robustness even for the drugs that vary significantly from these used during training, both in terms of chemical structure, drug class, and the year of FDA approval. AnimalGAN showed comparable results in hepatotoxicity assessment as using the real animal data and outperformed 12 conventional quantitative structure-activity relationship approaches. Using AnimalGAN, a virtual experiment of 100,000 rats ranked hepatotoxicity of three structurally similar drugs in a similar trend that has been observed in human population. AnimalGAN represented a significant step with artificial intelligence towards the global effort in replacement, reduction, and refinement (3Rs) of animal use.
Collapse
Affiliation(s)
- Xi Chen
- National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Ruth Roberts
- ApconiX Ltd, Alderley Park, Alderley Edge, SK10 4TG, UK
- University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Zhichao Liu
- National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR, 72079, USA.
- Currently working at Integrative Toxicology, Nonclinical Drug Safety, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, 06877, USA.
| | - Weida Tong
- National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR, 72079, USA.
| |
Collapse
|
18
|
Hu C, Yang W. Alternatives to animal models to study bacterial infections. Folia Microbiol (Praha) 2023; 68:703-739. [PMID: 37632640 DOI: 10.1007/s12223-023-01084-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 08/02/2023] [Indexed: 08/28/2023]
Abstract
Animal testing has made a significant and unequalled contribution to important discoveries and advancements in the fields of research, medicine, vaccine development, and drug discovery. Each year, millions of animals are sacrificed for various experiments, and this is an ongoing process. However, the debate on the ethical and sensible usage of animals in in vivo experimentation is equally important. The need to explore and adopt newer alternatives to animals so as to comply with the goal of reduce, refine, and replace needs attention. Besides the ever-increasing debate on ethical issues, animal research has additional drawbacks (need of trained labour, requirement of breeding area, lengthy protocols, high expenses, transport barriers, difficulty to extrapolate data from animals to humans, etc.). With this scenario, the present review has been framed to give a comprehensive insight into the possible alternative options worth exploring in this direction especially targeting replacements for animal models of bacterial infections. There have been some excellent reviews discussing on the alternate methods for replacing and reducing animals in drug research. However, reviews that discuss the replacements in the field of medical bacteriology with emphasis on animal bacterial infection models are purely limited. The present review discusses on the use of (a) non-mammalian models and (b) alternative systems such as microfluidic chip-based models and microdosing aiming to give a detailed insight into the prospects of these alternative platforms to reduce the number of animals being used in infection studies. This would enlighten the scientific community working in this direction to be well acquainted with the available new approaches and alternatives so that the 3R strategy can be successfully implemented in the field of antibacterial drug research and testing.
Collapse
Affiliation(s)
- Chengming Hu
- Queen Mary College, Nanchang University, Nanchang, China
| | - Wenlong Yang
- Department of Infectious Diseases, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
19
|
Lejal V, Cerisier N, Rouquié D, Taboureau O. Assessment of Drug-Induced Liver Injury through Cell Morphology and Gene Expression Analysis. Chem Res Toxicol 2023; 36:1456-1470. [PMID: 37652439 PMCID: PMC10523580 DOI: 10.1021/acs.chemrestox.2c00381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Indexed: 09/02/2023]
Abstract
Drug-induced liver injury (DILI) is a significant concern in drug development, often leading to drug withdrawal. Although many studies aim to identify biomarkers and gene/pathway signatures related to liver toxicity and aim to predict DILI compounds, this remains a challenge in drug discovery. With a strong development of high-content screening/imaging (HCS/HCI) for phenotypic screening, we explored the morphological cell perturbations induced by DILI compounds. In the first step, cell morphological signatures were associated with two datasets of DILI chemicals (DILIRank and eTox). The mechanisms of action were then analyzed for chemicals having transcriptomics data and sharing similar morphological perturbations. Signaling pathways associated with liver toxicity (cell cycle, cell growth, apoptosis, ...) were then captured, and a hypothetical relation between cell morphological perturbations and gene deregulation was illustrated within our analysis. Finally, using the cell morphological signatures, machine learning approaches were developed to predict chemicals with a potential risk of DILI. Some models showed relevant performance with validation set balanced accuracies between 0.645 and 0.739. Overall, our findings demonstrate the utility of combining HCI with transcriptomics data to identify the morphological and gene expression signatures related to DILI chemicals. Moreover, our protocol could be extended to other toxicity end points, offering a promising avenue for comprehensive toxicity assessment in drug discovery.
Collapse
Affiliation(s)
- Vanille Lejal
- Université
Paris Cité, Inserm U1133, CNRS
UMR 8251, 75013, Paris, France
| | - Natacha Cerisier
- Université
Paris Cité, Inserm U1133, CNRS
UMR 8251, 75013, Paris, France
| | - David Rouquié
- Bayer
SAS, Bayer Crop Science, 355 rue Dostoïevski, CS 90153, 06906 Valbonne, Sophia-Antipolis, France
- Université
Côte d’Azur 3IA Interdisciplinary Institute in Artificial Intelligence, 06103 Nice Cedex, France
| | - Olivier Taboureau
- Université
Paris Cité, Inserm U1133, CNRS
UMR 8251, 75013, Paris, France
| |
Collapse
|
20
|
Shin HK, Huang R, Chen M. In silico modeling-based new alternative methods to predict drug and herb-induced liver injury: A review. Food Chem Toxicol 2023; 179:113948. [PMID: 37460037 PMCID: PMC10640386 DOI: 10.1016/j.fct.2023.113948] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/10/2023] [Accepted: 07/14/2023] [Indexed: 07/25/2023]
Abstract
New approach methods (NAMs) have been developed to predict a wide range of toxicities through innovative technologies. Liver injury is one of the most extensively studied endpoints due to its severity and frequency, occurring among populations that consume drugs or dietary supplements. In this review, we focus on recent developments of in silico modeling for liver injury prediction using deep learning and in vitro data based on adverse outcome pathways (AOPs). Despite these models being mainly developed using datasets generated from drug-like molecules, they were also applied to the prediction of hepatotoxicity caused by herbal products. As deep learning has achieved great success in many different fields, advanced machine learning algorithms have been actively applied to improve the accuracy of in silico models. Additionally, the development of liver AOPs, combined with big data in toxicology, has been valuable in developing in silico models with enhanced predictive performance and interpretability. Specifically, one approach involves developing structure-based models for predicting molecular initiating events of liver AOPs, while others use in vitro data with structure information as model inputs for making predictions. Even though liver injury remains a difficult endpoint to predict, advancements in machine learning algorithms and the expansion of in vitro databases with relevant biological knowledge have made a huge impact on improving in silico modeling for drug-induced liver injury prediction.
Collapse
Affiliation(s)
- Hyun Kil Shin
- Department of Predictive Toxicology, Korea Institute of Toxicology (KIT), 34114, Daejeon, Republic of Korea
| | - Ruili Huang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, 20850, USA.
| | - Minjun Chen
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration, 3900 NCTR Rd., Jefferson, AR, 72079, USA.
| |
Collapse
|
21
|
Bussola N, Xu J, Wu L, Gorini L, Zhang Y, Furlanello C, Tong W. A Weakly Supervised Deep Learning Framework for Whole Slide Classification to Facilitate Digital Pathology in Animal Study. Chem Res Toxicol 2023; 36:1321-1331. [PMID: 37540590 PMCID: PMC10445282 DOI: 10.1021/acs.chemrestox.3c00058] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Indexed: 08/06/2023]
Abstract
The pathology of animal studies is crucial for toxicity evaluations and regulatory assessments, but the manual examination of slides by pathologists remains time-consuming and requires extensive training. One inherent challenge in this process is the interobserver variability, which can compromise the consistency and accuracy of a study. Artificial intelligence (AI) has demonstrated its ability to automate similar examinations in clinical applications with enhanced efficiency, consistency, and accuracy. However, training AI models typically relies on costly pixel-level annotation of injured regions and is often not available for animal pathology. To address this, we developed the PathologAI system, a "weakly" supervised approach for WSI classification in rat images without explicit lesion annotation at the pixel level. Using rat liver imaging data from the Open TG-GATEs system, PathologAI was applied to predict necrosis of n = 816 WSIs (377 controls). TG-GATEs studied 170 compounds at three dose levels (low, middle, and high) for four time points (3, 7, 14, and 28 days). PathologAI first preprocessed WSIs at the tile level to generate a high-level representation with a Generative Adversarial Network architecture. The prediction of liver necrosis relied on an ensemble model of 5 CNN classifiers trained on 335 WSIs. The ensemble model achieved notable classification accuracy on the holdout test set: 87% among 87 control slides free of findings, 83% among 120 controls with spontaneous necrosis, 67% among 147 treated animals with spontaneous minimal or slight necrosis, and 59% among 127 treated animals with minimal or slight necrosis caused by the treatment. Importantly, PathologAI was able to discriminate WSIs with spontaneous necrosis from those with treatment related necrosis and discriminated mild lesion level findings (slight vs minimal) and between treatment dose levels. PathologAI could provide an inexpensive and rapid screening tool to assist the digital pathology analysis in preclinical applications and general toxicological studies.
Collapse
Affiliation(s)
- Nicole Bussola
- Center
for Integrative Biology, University of Trento, Trento 38123, Italy
| | - Joshua Xu
- Division
of Bioinformatics and Biostatistics National
Center for Toxicological Research, Food
and Drug Administration, Jefferson, Arkansas 72079, United States
| | - Leihong Wu
- Division
of Bioinformatics and Biostatistics National
Center for Toxicological Research, Food
and Drug Administration, Jefferson, Arkansas 72079, United States
| | | | - Yifan Zhang
- Division
of Bioinformatics and Biostatistics National
Center for Toxicological Research, Food
and Drug Administration, Jefferson, Arkansas 72079, United States
| | | | - Weida Tong
- Division
of Bioinformatics and Biostatistics National
Center for Toxicological Research, Food
and Drug Administration, Jefferson, Arkansas 72079, United States
| |
Collapse
|
22
|
Li T, Roberts R, Liu Z, Tong W. TransOrGAN: An Artificial Intelligence Mapping of Rat Transcriptomic Profiles between Organs, Ages, and Sexes. Chem Res Toxicol 2023; 36:916-925. [PMID: 37200521 PMCID: PMC10433534 DOI: 10.1021/acs.chemrestox.3c00037] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Indexed: 05/20/2023]
Abstract
Animal studies are required for the evaluation of candidate drugs to ensure patient and volunteer safety. Toxicogenomics is often applied in these studies to gain understanding of the underlying mechanisms of toxicity, which is usually focused on critical organs such as the liver or kidney in young male rats. There is a strong ethical reason to reduce, refine and replace animal use (the 3Rs), where the mapping of data between organs, sexes and ages could reduce the cost and time of drug development. Herein, we proposed a generative adversarial network (GAN)-based framework entitled TransOrGAN that allowed the molecular mapping of gene expression profiles in different rodent organ systems and across sex and age groups. We carried out a proof-of-concept study based on rat RNA-seq data from 288 samples in 9 different organs of both sexes and 4 developmental stages. First, we demonstrated that TransOrGAN could infer transcriptomic profiles between any 2 of the 9 organs studied, yielding an average cosine similarity of 0.984 between synthetic transcriptomic profiles and their corresponding real profiles. Second, we found that TransOrGAN could infer transcriptomic profiles observed in females from males, with an average cosine similarity of 0.984. Third, we found that TransOrGAN could infer transcriptomic profiles in juvenile, adult, and aged animals from adolescent animals with an average cosine similarity of 0.981, 0.983, and 0.989, respectively. Altogether, TransOrGAN is an innovative approach to infer transcriptomic profiles between ages, sexes, and organ systems, offering the opportunity to reduce animal usage and to provide an integrated assessment of toxicity in the whole organism irrespective of sex or age.
Collapse
Affiliation(s)
- Ting Li
- National
Center for Toxicological Research, Food
and Drug Administration, Jefferson, Arkansas 72079, United States
| | - Ruth Roberts
- ApconiX Ltd, Alderley Park, Alderley Edge SK10 4TG, United Kingdom
- University
of Birmingham, Edgbaston, Birmingham B15 2TT, United
Kingdom
| | - Zhichao Liu
- Integrative
Toxicology, Nonclinical Drug Safety, Boehringer
Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - Weida Tong
- National
Center for Toxicological Research, Food
and Drug Administration, Jefferson, Arkansas 72079, United States
| |
Collapse
|
23
|
Rao M, Nassiri V, Alhambra C, Snoeys J, Van Goethem F, Irrechukwu O, Aleo MD, Geys H, Mitra K, Will Y. AI/ML Models to Predict the Severity of Drug-Induced Liver Injury for Small Molecules. Chem Res Toxicol 2023. [PMID: 37294641 DOI: 10.1021/acs.chemrestox.3c00098] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Drug-induced liver injury (DILI), believed to be a multifactorial toxicity, has been a leading cause of attrition of small molecules during discovery, clinical development, and postmarketing. Identification of DILI risk early reduces the costs and cycle times associated with drug development. In recent years, several groups have reported predictive models that use physicochemical properties or in vitro and in vivo assay endpoints; however, these approaches have not accounted for liver-expressed proteins and drug molecules. To address this gap, we have developed an integrated artificial intelligence/machine learning (AI/ML) model to predict DILI severity for small molecules using a combination of physicochemical properties and off-target interactions predicted in silico. We compiled a data set of 603 diverse compounds from public databases. Among them, 164 were categorized as Most DILI (M-DILI), 245 as Less DILI (L-DILI), and 194 as No DILI (N-DILI) by the FDA. Six machine learning methods were used to create a consensus model for predicting the DILI potential. These methods include k-nearest neighbor (k-NN), support vector machine (SVM), random forest (RF), Naïve Bayes (NB), artificial neural network (ANN), logistic regression (LR), weighted average ensemble learning (WA) and penalized logistic regression (PLR). Among the analyzed ML methods, SVM, RF, LR, WA, and PLR identified M-DILI and N-DILI compounds, achieving a receiver operating characteristic area under the curve of 0.88, sensitivity of 0.73, and specificity of 0.9. Approximately 43 off-targets, along with physicochemical properties (fsp3, log S, basicity, reactive functional groups, and predicted metabolites), were identified as significant factors in distinguishing between M-DILI and N-DILI compounds. The key off-targets that we identified include: PTGS1, PTGS2, SLC22A12, PPARγ, RXRA, CYP2C9, AKR1C3, MGLL, RET, AR, and ABCC4. The present AI/ML computational approach therefore demonstrates that the integration of physicochemical properties and predicted on- and off-target biological interactions can significantly improve DILI predictivity compared to chemical properties alone.
Collapse
Affiliation(s)
- Mohan Rao
- Discovery, Product Development and Supply (DPDS), Preclinical Sciences and Translational Safety (PSTS), Predictive Investigative and Translational Toxicology (PITT), Janssen Pharmaceutical Companies of Johnson and Johnson, La Jolla, California 92121, United States
| | - Vahid Nassiri
- Open Analytics, Jupiterstraat 20, 2600 Antwerpen, Belgium
| | - Cristóbal Alhambra
- Discovery, Product Development and Supply (DPDS), Preclinical Sciences and Translational Safety (PSTS), Predictive Investigative and Translational Toxicology (PITT), Janssen Pharmaceutical Companies of Johnson and Johnson, La Jolla, California 92121, United States
| | - Jan Snoeys
- Discovery, Product Development and Supply (DPDS), Preclinical Sciences and Translational Safety (PSTS), Predictive Investigative and Translational Toxicology (PITT), Janssen Pharmaceutical Companies of Johnson and Johnson, La Jolla, California 92121, United States
| | - Freddy Van Goethem
- Discovery, Product Development and Supply (DPDS), Preclinical Sciences and Translational Safety (PSTS), Predictive Investigative and Translational Toxicology (PITT), Janssen Pharmaceutical Companies of Johnson and Johnson, La Jolla, California 92121, United States
| | - Onyi Irrechukwu
- Discovery, Product Development and Supply (DPDS), Preclinical Sciences and Translational Safety (PSTS), Predictive Investigative and Translational Toxicology (PITT), Janssen Pharmaceutical Companies of Johnson and Johnson, La Jolla, California 92121, United States
| | - Michael D Aleo
- TOXinsights LLC, Boiling Springs, Pennsylvania 17007, United States
| | - Helena Geys
- Discovery, Product Development and Supply (DPDS), Preclinical Sciences and Translational Safety (PSTS), Predictive Investigative and Translational Toxicology (PITT), Janssen Pharmaceutical Companies of Johnson and Johnson, La Jolla, California 92121, United States
| | - Kaushik Mitra
- Discovery, Product Development and Supply (DPDS), Preclinical Sciences and Translational Safety (PSTS), Predictive Investigative and Translational Toxicology (PITT), Janssen Pharmaceutical Companies of Johnson and Johnson, La Jolla, California 92121, United States
| | - Yvonne Will
- Discovery, Product Development and Supply (DPDS), Preclinical Sciences and Translational Safety (PSTS), Predictive Investigative and Translational Toxicology (PITT), Janssen Pharmaceutical Companies of Johnson and Johnson, La Jolla, California 92121, United States
| |
Collapse
|
24
|
Nault R, Cave MC, Ludewig G, Moseley HN, Pennell KG, Zacharewski T. A Case for Accelerating Standards to Achieve the FAIR Principles of Environmental Health Research Experimental Data. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:65001. [PMID: 37352010 PMCID: PMC10289218 DOI: 10.1289/ehp11484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/25/2023]
Abstract
BACKGROUND Funding agencies, publishers, and other stakeholders are pushing environmental health science investigators to improve data sharing; to promote the findable, accessible, interoperable, and reusable (FAIR) principles; and to increase the rigor and reproducibility of the data collected. Accomplishing these goals will require significant cultural shifts surrounding data management and strategies to develop robust and reliable resources that bridge the technical challenges and gaps in expertise. OBJECTIVE In this commentary, we examine the current state of managing data and metadata-referred to collectively as (meta)data-in the experimental environmental health sciences. We introduce new tools and resources based on in vivo experiments to serve as examples for the broader field. METHODS We discuss previous and ongoing efforts to improve (meta)data collection and curation. These include global efforts by the Functional Genomics Data Society to develop metadata collection tools such as the Investigation, Study, Assay (ISA) framework, and the Center for Expanded Data Annotation and Retrieval. We also conduct a case study of in vivo data deposited in the Gene Expression Omnibus that demonstrates the current state of in vivo environmental health data and highlights the value of using the tools we propose to support data deposition. DISCUSSION The environmental health science community has played a key role in efforts to achieve the goals of the FAIR guiding principles and is well positioned to advance them further. We present a proposed framework to further promote these objectives and minimize the obstacles between data producers and data scientists to maximize the return on research investments. https://doi.org/10.1289/EHP11484.
Collapse
Affiliation(s)
- Rance Nault
- Biochemistry & Molecular Biology Department, Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Matthew C. Cave
- Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville, Louisville, Kentucky, USA
| | - Gabriele Ludewig
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, Iowa, USA
| | - Hunter N.B. Moseley
- Molecular and Cellular Biochemistry Department, University of Kentucky, Lexington, Kentucky, USA
| | - Kelly G. Pennell
- Department of Civil Engineering, University of Kentucky, Lexington, Kentucky, USA
| | - Tim Zacharewski
- Biochemistry & Molecular Biology Department, Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
25
|
Naciff JM, Shan YK, Wang X, Daston GP. Article title: Transcriptional profiling efficacy to define biological activity similarity for cosmetic ingredients' safety assessment based on next-generation read-across. FRONTIERS IN TOXICOLOGY 2022; 4:1082222. [PMID: 36618549 PMCID: PMC9811170 DOI: 10.3389/ftox.2022.1082222] [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: 10/27/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
The objective of this work was to use transcriptional profiling to assess the biological activity of structurally related chemicals to define their biological similarity and with that, substantiate the validity of a read-across approach usable in risk assessment. Two case studies are presented, one with 4 short alkyl chain parabens: methyl (MP), ethyl (EP), butyl (BP), and propylparaben (PP), as well as their main metabolite, p-hydroxybenzoic acid (pHBA) with the assumption that propylparaben was the target chemical; and a second one with caffeine and its main metabolites theophylline, theobromine and paraxanthine where CA was the target chemical. The comprehensive transcriptional response of MCF7, HepG2, A549 and ICell cardiomyocytes was evaluated (TempO-Seq) after exposure to vehicle-control, each paraben or pHBA, CA or its metabolites, at 3 non-cytotoxic concentrations, for 6 h. Differentially expressed genes (FDR ≥0.05, and fold change ±1.2≥) were identified for each chemical, at each concentration, and used to determine similarities. Each of the chemicals is able to elicit changes in the expression of a number of genes, as compared to controls. Importantly, the transcriptional profile elicited by each of the parabens shares a high degree of similarity across the group. The highest number of genes commonly affected was between butylparaben and PP. The transcriptional profile of the parabens is similar to the one elicited by estrogen receptor agonists, with BP being the closest structural and biological analogue for PP. In the CA case, the transcriptional profile elicited of all four methylxanthines had a high degree of similarity across the cell types, with CA and theophylline being the most active. The most robust response was obtained in the cardiomyocytes with the highest transcriptional profile similarity between CA and TP. The transcriptional profile of the methylxanthines is similar to the one elicited by inhibitors of phosphatidylinositol 3-kinase as well as other kinase inhibitors. Overall, our results support the approach of incorporating transcriptional profiling in well-designed in vitro tests as one robust stream of data to support biological similarity driven read-across procedures and strengthening the traditional structure-based approaches useful in risk assessment.
Collapse
|
26
|
Sniatynski MJ, Kristal BS. Predicting drug toxicity at the intersection of informatics and biology: DTox builds a foundation. PATTERNS (NEW YORK, N.Y.) 2022; 3:100586. [PMID: 36124303 PMCID: PMC9481942 DOI: 10.1016/j.patter.2022.100586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Hao et al. (2022) present DTox (deep learning for toxicology), a neural network designed to predict and probe the sites and potential mechanisms underlying chemical toxicity; results provide a map to facilitate modular testing and improvements across multiple disparate applications.
Collapse
Affiliation(s)
- Matthew J. Sniatynski
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital, 221 Longwood Ave, LM322B, Boston, MA 02115, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Bruce S. Kristal
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital, 221 Longwood Ave, LM322B, Boston, MA 02115, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
27
|
Abstract
Machine learning and artificial intelligence approaches have revolutionized multiple disciplines, including toxicology. This review summarizes representative recent applications of machine learning and artificial intelligence approaches in different areas of toxicology, including physiologically based pharmacokinetic (PBPK) modeling, quantitative structure-activity relationship modeling for toxicity prediction, adverse outcome pathway analysis, high-throughput screening, toxicogenomics, big data and toxicological databases. By leveraging machine learning and artificial intelligence approaches, now it is possible to develop PBPK models for hundreds of chemicals efficiently, to create in silico models to predict toxicity for a large number of chemicals with similar accuracies compared to in vivo animal experiments, and to analyze a large amount of different types of data (toxicogenomics, high-content image data, etc.) to generate new insights into toxicity mechanisms rapidly, which was impossible by manual approaches in the past. To continue advancing the field of toxicological sciences, several challenges should be considered: (1) not all machine learning models are equally useful for a particular type of toxicology data, and thus it is important to test different methods to determine the optimal approach; (2) current toxicity prediction is mainly on bioactivity classification (yes/no), so additional studies are needed to predict the intensity of effect or dose-response relationship; (3) as more data become available, it is crucial to perform rigorous data quality check and develop infrastructure to store, share, analyze, evaluate, and manage big data; and (4) it is important to convert machine learning models to user-friendly interfaces to facilitate their applications by both computational and bench scientists.
Collapse
Affiliation(s)
- Zhoumeng Lin
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL, 32610, USA.,Center for Environmental and Human Toxicology, University of Florida, FL, 32608, USA
| | - Wei-Chun Chou
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL, 32610, USA.,Center for Environmental and Human Toxicology, University of Florida, FL, 32608, USA
| |
Collapse
|
28
|
Jeong J, Choi J. Artificial Intelligence-Based Toxicity Prediction of Environmental Chemicals: Future Directions for Chemical Management Applications. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:7532-7543. [PMID: 35666838 DOI: 10.1021/acs.est.1c07413] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Recently, research on the development of artificial intelligence (AI)-based computational toxicology models that predict toxicity without the use of animal testing has emerged because of the rapid development of computer technology. Various computational toxicology techniques that predict toxicity based on the structure of chemical substances are gaining attention, including the quantitative structure-activity relationship. To understand the recent development of these models, we analyzed the databases, molecular descriptors, fingerprints, and algorithms considered in recent studies. Based on a selection of 96 papers published since 2014, we found that AI models have been developed to predict approximately 30 different toxicity end points using more than 20 toxicity databases. For model development, molecular access system and extended-connectivity fingerprints are the most commonly used molecular descriptors. The most used algorithm among the machine learning techniques is the random forest, while the most used algorithm among the deep learning techniques is a deep neural network. The use of AI technology in the development of toxicity prediction models is a new concept that will aid in achieving a scientific accord and meet regulatory applications. The comprehensive overview provided in this study will provide a useful guide for the further development and application of toxicity prediction models.
Collapse
Affiliation(s)
- Jaeseong Jeong
- School of Environmental Engineering, University of Seoul, 163 Seoulsiripdae-ro, Dongdaemun-gu, Seoul 02504, South Korea
| | - Jinhee Choi
- School of Environmental Engineering, University of Seoul, 163 Seoulsiripdae-ro, Dongdaemun-gu, Seoul 02504, South Korea
| |
Collapse
|
29
|
Shin HK, Florean O, Hardy B, Doktorova T, Kang MG. Semi-automated approach for generation of biological networks on drug-induced cholestasis, steatosis, hepatitis, and cirrhosis. Toxicol Res 2022; 38:393-407. [PMID: 35865277 PMCID: PMC9247124 DOI: 10.1007/s43188-022-00124-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/02/2022] [Accepted: 02/11/2022] [Indexed: 12/03/2022] Open
Abstract
Drug-induced liver injury (DILI) is one of the leading reasons for discontinuation of a new drug development project. Diverse machine learning or deep learning models have been developed to predict DILI. However, these models have not provided an adequate understanding of the mechanisms leading to DILI. The development of safer drugs requires novel computational approaches that enable the prompt understanding of the mechanism of DILI. In this study, the mechanisms leading to the development of cholestasis, steatosis, hepatitis, and cirrhosis were explored using a semi-automated approach for data gathering and associations. Diverse data from ToxCast, Comparative Toxicogenomic Database (CTD), Reactome, and Open TG-GATEs on reference molecules leading to the development of the respective diseases were extracted. The data were used to create biological networks of the four diseases. As expected, the four networks had several common pathways, and a joint DILI network was assembled. Such biological networks could be used in drug discovery to identify possible molecules of concern as they provide a better understanding of the disease-specific key events. The events can be target-tested to provide indications for potential DILI effects.
Collapse
Affiliation(s)
- Hyun Kil Shin
- Toxicoinformatics Group, Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 34114 Republic of Korea
- Human and Environmental Toxicology, University of Science and Technology, Daejeon, 34113 Republic of Korea
| | - Oana Florean
- Edelweiss Connect GmbH, Hochbergerstrasse 60C, 4057 Basel, Switzerland
| | - Barry Hardy
- Edelweiss Connect GmbH, Hochbergerstrasse 60C, 4057 Basel, Switzerland
| | - Tatyana Doktorova
- Edelweiss Connect GmbH, Hochbergerstrasse 60C, 4057 Basel, Switzerland
| | - Myung-Gyun Kang
- Toxicoinformatics Group, Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 34114 Republic of Korea
| |
Collapse
|