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
|
Maciag M, Karamyan VT. The Missing Enzymes: A Call to Update Pharmacological Profiling Practices for Better Drug Safety Assessment. J Med Chem 2025; 68:7854-7865. [PMID: 40173276 PMCID: PMC12035801 DOI: 10.1021/acs.jmedchem.4c02228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 01/14/2025] [Accepted: 03/21/2025] [Indexed: 04/04/2025]
Abstract
Pharmacological profiling is critical for the development of safe drugs. With increasing awareness of its significance and attempts to share best practices, here we aimed to understand how pharmacological profiling is implemented and reported in the primary literature by analyzing the representation of nonkinase enzymes in selectivity screens. This aspect has been overlooked in previous publications, despite enzymes constituting a significant portion of the pharmacological targets for currently marketed drugs. Our analysis shows that while industry recommendations for improved pharmacological profiling have been widely adopted, enzymes remain largely underrepresented: about a quarter of studies did not include enzymes, and on average, enzymes comprise only 11% of all targets in pharmacological screens. We discuss possible reasons for this shortcoming and provide examples of critical enzymes missing from current screens. We conclude with the notion that selectivity screens should be expanded to include more enzymes to improve drug development and safety.
Collapse
Affiliation(s)
- Monika Maciag
- Department of Foundational Medical
Studies, William Beaumont School of Medicine, Oakland University, Rochester, Michigan 48309, United States
| | - Vardan T. Karamyan
- Department of Foundational Medical
Studies, William Beaumont School of Medicine, Oakland University, Rochester, Michigan 48309, United States
| |
Collapse
|
3
|
Tanoli Z, Fernández-Torras A, Özcan UO, Kushnir A, Nader KM, Gadiya Y, Fiorenza L, Ianevski A, Vähä-Koskela M, Miihkinen M, Seemab U, Leinonen H, Seashore-Ludlow B, Tampere M, Kalman A, Ballante F, Benfenati E, Saunders G, Potdar S, Gómez García I, García-Serna R, Talarico C, Beccari AR, Schaal W, Polo A, Costantini S, Cabri E, Jacobs M, Saarela J, Budillon A, Spjuth O, Östling P, Xhaard H, Quintana J, Mestres J, Gribbon P, Ussi AE, Lo DC, de Kort M, Wennerberg K, Fratelli M, Carreras-Puigvert J, Aittokallio T. Computational drug repurposing: approaches, evaluation of in silico resources and case studies. Nat Rev Drug Discov 2025:10.1038/s41573-025-01164-x. [PMID: 40102635 DOI: 10.1038/s41573-025-01164-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2025] [Indexed: 03/20/2025]
Abstract
Repurposing of existing drugs for new indications has attracted substantial attention owing to its potential to accelerate drug development and reduce costs. Hundreds of computational resources such as databases and predictive platforms have been developed that can be applied for drug repurposing, making it challenging to select the right resource for a specific drug repurposing project. With the aim of helping to address this challenge, here we overview computational approaches to drug repurposing based on a comprehensive survey of available in silico resources using a purpose-built drug repurposing ontology that classifies the resources into hierarchical categories and provides application-specific information. We also present an expert evaluation of selected resources and three drug repurposing case studies implemented within the Horizon Europe REMEDi4ALL project to demonstrate the practical use of the resources. This comprehensive Review with expert evaluations and case studies provides guidelines and recommendations on the best use of various in silico resources for drug repurposing and establishes a basis for a sustainable and extendable drug repurposing web catalogue.
Collapse
Affiliation(s)
- Ziaurrehman Tanoli
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Drug Discovery and Chemical Biology (DDCB) Consortium, Biocenter Finland, University of Helsinki, Helsinki, Finland.
| | | | - Umut Onur Özcan
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Aleksandr Kushnir
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Kristen Michelle Nader
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Yojana Gadiya
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Frankfurt, Germany
- Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, Bonn, Germany
| | - Laura Fiorenza
- Dipartimento di Elettronica, Informazione e Bioingegneria (DEIB), Politecnico di Milano, Milan, Italy
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Aleksandr Ianevski
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Markus Vähä-Koskela
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mitro Miihkinen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Umair Seemab
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Henri Leinonen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Brinton Seashore-Ludlow
- Science for Life Laboratory (SciLifeLab), Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Marianna Tampere
- Science for Life Laboratory (SciLifeLab), Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Adelinn Kalman
- Science for Life Laboratory (SciLifeLab), Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Flavio Ballante
- Chemical Biology Consortium Sweden (CBCS), SciLifeLab, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Emilio Benfenati
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Gary Saunders
- European Infrastructure for Translational Medicine (EATRIS ERIC), Amsterdam, The Netherlands
| | - Swapnil Potdar
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | | | | | | | | | - Wesley Schaal
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Andrea Polo
- Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Susan Costantini
- Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Enrico Cabri
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marc Jacobs
- Fraunhofer-Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, Germany
| | - Jani Saarela
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Alfredo Budillon
- Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Ola Spjuth
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Päivi Östling
- Science for Life Laboratory (SciLifeLab), Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Henri Xhaard
- Drug Discovery and Chemical Biology (DDCB) Consortium, Biocenter Finland, University of Helsinki, Helsinki, Finland
- Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Jordi Quintana
- Chemotargets SL, Parc Científic de Barcelona, Barcelona, Catalonia, Spain
| | - Jordi Mestres
- Chemotargets SL, Parc Científic de Barcelona, Barcelona, Catalonia, Spain
- Institut de Quimica Computacional i Catalisi, Facultat de Ciencies, Universitat de Girona, Girona, Catalonia, Spain
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Frankfurt, Germany
| | - Anton E Ussi
- European Infrastructure for Translational Medicine (EATRIS ERIC), Amsterdam, The Netherlands
| | - Donald C Lo
- European Infrastructure for Translational Medicine (EATRIS ERIC), Amsterdam, The Netherlands
| | - Martin de Kort
- European Infrastructure for Translational Medicine (EATRIS ERIC), Amsterdam, The Netherlands
| | - Krister Wennerberg
- Biotech Research & Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | | | - Jordi Carreras-Puigvert
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway.
- Oslo Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
4
|
Creanza TM, Alberga D, Patruno C, Mangiatordi GF, Ancona N. Transformer Decoder Learns from a Pretrained Protein Language Model to Generate Ligands with High Affinity. J Chem Inf Model 2025; 65:1258-1277. [PMID: 39871540 DOI: 10.1021/acs.jcim.4c02019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
The drug discovery process can be significantly accelerated by using deep learning methods to suggest molecules with druglike features and, more importantly, that are good candidates to bind specific proteins of interest. We present a novel deep learning generative model, Prot2Drug, that learns to generate ligands binding specific targets leveraging (i) the information carried by a pretrained protein language model and (ii) the ability of transformers to capitalize the knowledge gathered from thousands of protein-ligand interactions. The embedding unveils the receipt to follow for designing molecules binding a given protein, and Prot2Drug translates such instructions by using the syntax of the molecular language generating novel compounds which are predicted to have favorable physicochemical properties and high affinity toward specific targets. Moreover, Prot2Drug reproduced numerous known interactions between compounds and proteins used for generating them and suggested novel protein targets for known compounds, indicating potential drug repurposing strategies. Remarkably, Prot2Drug facilitates the design of promising ligands even for protein targets with limited or no information about their ligands or 3D structure.
Collapse
Affiliation(s)
- Teresa Maria Creanza
- Institute of Intelligent Industrial Technologies and Systems for Advanced Manufacturing, Consiglio Nazionale delle Ricerche, Via G. Amendola, 122/d, Bari 70126, Italy
| | - Domenico Alberga
- Institute of Crystallography, Consiglio Nazionale delle Ricerche, Via G. Amendola, 122/d, Bari 70126, Italy
| | - Cosimo Patruno
- Institute of Intelligent Industrial Technologies and Systems for Advanced Manufacturing, Consiglio Nazionale delle Ricerche, Via G. Amendola, 122/d, Bari 70126, Italy
| | | | - Nicola Ancona
- Institute of Intelligent Industrial Technologies and Systems for Advanced Manufacturing, Consiglio Nazionale delle Ricerche, Via G. Amendola, 122/d, Bari 70126, Italy
| |
Collapse
|
5
|
Rouseti GM, Fischer A, Rathfelder N, Grimes K, Waldt A, Cuttat R, Schuierer S, Wild S, Jivkov M, Dubost V, Schadt HS, Odermatt A, Vicart A, Moretti F. Disruption of serotonin homeostasis in intestinal organoids provides insights into drug-induced gastrointestinal toxicity. Toxicology 2025; 511:154028. [PMID: 39643203 DOI: 10.1016/j.tox.2024.154028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/25/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Drug-induced gastrointestinal toxicity is a frequent clinical adverse event that needs to be carefully monitored and managed to ensure patient compliance. While preclinical assessment of drug-induced gastrointestinal toxicity mostly relies on animal experimentation, intestinal organoids have gained increasing attention to identify gastrointestinal toxicants in vitro. Nonetheless, current in vitro protocols primarily assess structural alterations induced by drugs, whereas gastrointestinal adverse events can often stem from functional disturbances. Disruption of serotonin signaling in the gastrointestinal tract is associated with impaired motility, as well as nausea and vomiting. We aimed to investigate alterations of serotonin homeostasis in organoids derived from the canine small intestine as a driver of drug-induced gastrointestinal toxicity. Treatment of the organoids with a compound (NVS-1) inducing acute gastrointestinal toxicity in dogs as well as with three tyrosine kinase inhibitors with known preclinical and clinical gastrointestinal adverse effects (afatinib, crizotinib and vandetanib) led to increased supernatant serotonin levels. Mechanistic assays showed that, while NVS-1 and afatinib stimulate serotonin release, crizotinib and vandetanib inhibit serotonin re-uptake via direct inhibition of the serotonin re-uptake transporter. Using a data mining approach, we further suggest that inhibition of serotonin re-uptake could contribute to gastrointestinal toxicity observed with multiple marketed drugs. In conclusion, we present the implementation of a novel in vitro gastrointestinal toxicity endpoint that could complement current methods and serve as a mechanistic and predictive/screening tool for drug-induced gastrointestinal toxicity.
Collapse
Affiliation(s)
- Georgia M Rouseti
- Preclinical Safety, Biomedical Research, Novartis Pharma AG, Basel, Switzerland; Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Audrey Fischer
- Preclinical Safety, Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Nicole Rathfelder
- Preclinical Safety, Biomedical Research, Novartis Pharma AG, Basel, Switzerland; present address: Department of Chemistry, University of Basel, Basel, Switzerland
| | - Karen Grimes
- Preclinical Safety, Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Annick Waldt
- Preclinical Safety, Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Rachel Cuttat
- Preclinical Safety, Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Sven Schuierer
- Preclinical Safety, Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Sophia Wild
- Preclinical Safety, Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Magali Jivkov
- Preclinical Safety, Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Valerie Dubost
- Preclinical Safety, Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Heiko S Schadt
- Preclinical Safety, Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland; Swiss Centre for Applied Human Toxicology and Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Axel Vicart
- Preclinical Safety, Biomedical Research, Novartis Pharma AG, Basel, Switzerland.
| | - Francesca Moretti
- Preclinical Safety, Biomedical Research, Novartis Pharma AG, Basel, Switzerland.
| |
Collapse
|
6
|
Redfern WS, Pollard CE, Holbrook M, Islam B, Abbasi M, Mahmud J, Lambert K, Haslam A, Jo H, Khalidi H, Bielecka Z, Starkey J, Ellinger T, Bryan S, Savas A, Andrews S, Aspbury R, Rosenbrier Ribeiro L, Henderson Park KA, Vargas HM, Gilmer CR. Predicting clinical outcomes from off-target receptor interactions using Secondary Intelligence™. J Pharmacol Toxicol Methods 2025; 131:107570. [PMID: 39577752 DOI: 10.1016/j.vascn.2024.107570] [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: 09/23/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
Adverse effects due to off-target activity can be predicted by careful comparison of the relationship between expected plasma concentration and off-target activity of the test compound with that of reference drugs targeting that receptor for their therapeutic efficacy. The ratio between plasma concentration (unbound) and the Ki at the receptor is a surrogate measure reflecting receptor occupancy. Where data are available for reference drugs, we have curated and evaluated this at 100 receptors, 72 of which can involve both negative and positive modulations by drugs: a total of 172 'receptor modulations'. This provides a quantitative framework upon which to achieve consistent risk assessment of off-target interactions across receptors, across compounds and between assessors. It therefore represents a significant departure from an opinion-based to an evidence-based approach to secondary pharmacology. Demonstration of proof-of-principle was achieved for one of the receptor interactions (α1A-adrenoceptor antagonism leading to postural hypotension in clinical use) due to the availability of high-quality off-target Ki data for >30 drugs at this receptor.
Collapse
Affiliation(s)
- Will S Redfern
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom.
| | - Chris E Pollard
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Mark Holbrook
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Barira Islam
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Mitra Abbasi
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Joanne Mahmud
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Katie Lambert
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Augustus Haslam
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Heeseung Jo
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Hiba Khalidi
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Zofia Bielecka
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom; Pharmacoepidemiology and Pharmacoeconomics Unit, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Josh Starkey
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Thomas Ellinger
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Simon Bryan
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Angeli Savas
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Steve Andrews
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Rob Aspbury
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | | | | | - Hugo M Vargas
- Translational Safety Research, TS&BA, Amgen Inc., Thousand Oaks, CA 91320, USA
| | - Clare R Gilmer
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| |
Collapse
|
7
|
Burbank M, Kukic P, Ouedraogo G, Kenna JG, Hewitt NJ, Armstrong D, Otto-Bruc A, Ebmeyer J, Boettcher M, Willox I, Mahony C. In vitro pharmacologic profiling aids systemic toxicity assessment of chemicals. Toxicol Appl Pharmacol 2024; 492:117131. [PMID: 39437896 DOI: 10.1016/j.taap.2024.117131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/02/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
An adapted in vitro pharmacology profiling panel (APPP) was developed that included targets used in the pharmaceutical industry alongside additional targets whose cellular functions have been linked to systemic toxicities. This panel of 83 target assays was used to profile the activities of 129 cosmetic relevant chemicals with diverse chemical structures, physiochemical properties and cosmetic use types. Internal data consistency was proved robust, as evidenced by the reproducibility between single concentration and concentration-response data and showed good concordance with data reported in the ToxCast and drug excipient datasets. We discuss how the data can be analyzed and multiple potential contexts of use illustrated by case studies, alongside other new approach methodologies, to support cosmetic chemical risk assessments that do not require data from new animal studies.
Collapse
Affiliation(s)
| | - Predrag Kukic
- Unilever Safety and Environmental Assurance Centre, Bedfordshire, MK 44 1LQ, UK
| | | | - J Gerry Kenna
- Cosmetics Europe, 40 Avenue Hermann-Debroux, 1160 Brussels, Belgium
| | - Nicola J Hewitt
- Cosmetics Europe, 40 Avenue Hermann-Debroux, 1160 Brussels, Belgium
| | | | | | | | | | - Ian Willox
- Eurofins Cerep, Celle-Lévescault, France
| | | |
Collapse
|
8
|
Popović L, Brankatschk B, Palladino G, Rossner MJ, Wehr MC. Polypharmacological profiling across protein target families and cellular pathways using the multiplexed cell-based assay platform safetyProfiler reveals efficacy, potency and side effects of drugs. Biomed Pharmacother 2024; 180:117523. [PMID: 39405910 DOI: 10.1016/j.biopha.2024.117523] [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: 08/16/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 11/14/2024] Open
Abstract
Selectivity profiling is key for assessing the pharmacological properties of multi-target drugs. We have developed a cell-based and barcoded assay encompassing ten druggable targets, including G protein-coupled receptors (GPCRs), receptor tyrosine kinases (RTKs), nuclear receptors, a protease as well as their key downstream pathways and profiled 17 drugs in living cells for efficacy, potency, and side effects. Notably, this multiplex assay, termed safetyProfiler assay, enabled the simultaneous assessment of multiple target and pathway activities, shedding light on the polypharmacological profile of compounds. For example, the neuroleptics clozapine, paliperidone, and risperidone potently inhibited primary targets DRD2 and HTR2A as well as cAMP and calcium pathways. However, while paliperidone and risperidone also potently inhibited the secondary target ADRA1A and mitogen-activated protein kinase (MAPK) downstream pathways, clozapine only exhibited mild antagonistic effects on ADRA1A and lacked MAPK inhibition downstream of DRD2 and HTR2A. Furthermore, we present data on the selectivity for bazedoxifene, an estrogen receptor antagonist currently undergoing clinical phase 2 trials for breast cancer, on MAPK signaling. Additionally, precise potency data for LY2452473, an androgen receptor antagonist, that completed a phase 2 clinical trial for prostate cancer, are presented. The non-selective kinase inhibitor staurosporine was observed to potently inactivate the two RTKs EGFR and ERBB4 as well as MAPK signaling, while eliciting stress-related cAMP responses. Our findings underscore the value of comprehensive profiling in elucidating the pharmacological properties of established and novel therapeutics, thereby facilitating the development of novel multi-target drugs with enhanced efficacy and selectivity.
Collapse
Affiliation(s)
- Lukša Popović
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstr. 7, Munich 80336, Germany; Systasy Bioscience GmbH, Fraunhoferstr. 8, Planegg-Martinsried 82152, Germany
| | - Ben Brankatschk
- Systasy Bioscience GmbH, Fraunhoferstr. 8, Planegg-Martinsried 82152, Germany
| | - Giulia Palladino
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstr. 7, Munich 80336, Germany; Systasy Bioscience GmbH, Fraunhoferstr. 8, Planegg-Martinsried 82152, Germany
| | - Moritz J Rossner
- Section of Molecular Neurobiology, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstr. 7, Munich 80336, Germany
| | - Michael C Wehr
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstr. 7, Munich 80336, Germany; Systasy Bioscience GmbH, Fraunhoferstr. 8, Planegg-Martinsried 82152, Germany.
| |
Collapse
|
9
|
Harmer AR, Rolf MG. On the relationship between hERG inhibition and the magnitude of QTc prolongation: An in vitro to clinical translational analysis. Toxicol Appl Pharmacol 2024; 492:117135. [PMID: 39490450 DOI: 10.1016/j.taap.2024.117135] [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: 09/16/2024] [Revised: 10/25/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
Assessing the magnitude of QTc prolongation is crucial in drug development due to its association with Torsades de Pointes. Inhibition of the hERG channel, pivotal in cardiac repolarization, is a key factor in evaluating this risk. In this study, the relationship between hERG inhibition and QTc prolongation magnitude was investigated, with the aim to derive simple guidance on the required hERG margin to avoid a large (>20 ms) QTc prolongation. METHODS Data from literature and FDA sources were searched for compounds with hERG IC50 values alongside clinical QTc data with paired plasma concentrations, or compounds demonstrating a clinical concentration-QTc relationship. Relationships between hERG inhibition, hERG IC50 margin to unbound plasma Cmax, and QTc prolongation magnitude were calculated. RESULTS Analysis of 148 clinical QTc observations from 98 compounds revealed that compounds associated with QTc prolongation >10 ms typically exhibited hERG margins of ≤33-fold, while those exceeding 20 ms were generally associated with margins of ≤24-fold. QTc increases above 10 ms were not observed at hERG margins >100-fold. Based on 53 clinical concentration-QTc datasets, modest hERG inhibition levels of ∼4-6 % correlated with a 10 ms QTc prolongation, while ∼10-13 % inhibition corresponded to a 20 ms prolongation. CONCLUSIONS This study enhances understanding of the relationship between hERG inhibition and QTc prolongation magnitude, by conducting analysis across a wide range of 98 compounds. This information can be used to determine the optimal hERG margin, particularly for drug discovery projects with limited scope to completely design-out hERG activity.
Collapse
Affiliation(s)
- Alexander R Harmer
- The Discovery Centre, Safety Sciences, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, UK.
| | - Michael G Rolf
- Gothenburg, Safety Sciences, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Sweden
| |
Collapse
|
10
|
Manen-Freixa L, Antolin AA. Polypharmacology prediction: the long road toward comprehensively anticipating small-molecule selectivity to de-risk drug discovery. Expert Opin Drug Discov 2024; 19:1043-1069. [PMID: 39004919 DOI: 10.1080/17460441.2024.2376643] [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: 03/15/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024]
Abstract
INTRODUCTION Small molecules often bind to multiple targets, a behavior termed polypharmacology. Anticipating polypharmacology is essential for drug discovery since unknown off-targets can modulate safety and efficacy - profoundly affecting drug discovery success. Unfortunately, experimental methods to assess selectivity present significant limitations and drugs still fail in the clinic due to unanticipated off-targets. Computational methods are a cost-effective, complementary approach to predict polypharmacology. AREAS COVERED This review aims to provide a comprehensive overview of the state of polypharmacology prediction and discuss its strengths and limitations, covering both classical cheminformatics methods and bioinformatic approaches. The authors review available data sources, paying close attention to their different coverage. The authors then discuss major algorithms grouped by the types of data that they exploit using selected examples. EXPERT OPINION Polypharmacology prediction has made impressive progress over the last decades and contributed to identify many off-targets. However, data incompleteness currently limits most approaches to comprehensively predict selectivity. Moreover, our limited agreement on model assessment challenges the identification of the best algorithms - which at present show modest performance in prospective real-world applications. Despite these limitations, the exponential increase of multidisciplinary Big Data and AI hold much potential to better polypharmacology prediction and de-risk drug discovery.
Collapse
Affiliation(s)
- Leticia Manen-Freixa
- Oncobell Division, Bellvitge Biomedical Research Institute (IDIBELL) and ProCURE Department, Catalan Institute of Oncology (ICO), Barcelona, Spain
| | - Albert A Antolin
- Oncobell Division, Bellvitge Biomedical Research Institute (IDIBELL) and ProCURE Department, Catalan Institute of Oncology (ICO), Barcelona, Spain
- Center for Cancer Drug Discovery, The Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| |
Collapse
|
11
|
Rao M, McDuffie E, Srivastava S, Plaisted W, Sachs C. Safety Implications of Modulating Nuclear Receptors: A Comprehensive Analysis from Non-Clinical and Clinical Perspectives. Pharmaceuticals (Basel) 2024; 17:875. [PMID: 39065726 PMCID: PMC11279859 DOI: 10.3390/ph17070875] [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: 05/09/2024] [Revised: 06/13/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
The unintended modulation of nuclear receptor (NR) activity by drugs can lead to toxicities amongst the endocrine, gastrointestinal, hepatic cardiovascular, and central nervous systems. While secondary pharmacology screening assays include NRs, safety risks due to unintended interactions of small molecule drugs with NRs remain poorly understood. To identify potential nonclinical and clinical safety effects resulting from functional interactions with 44 of the 48 human-expressed NRs, we conducted a systematic narrative review of the scientific literature, tissue expression data, and used curated databases (OFF-X™) (Off-X, Clarivate) to organize reported toxicities linked to the functional modulation of NRs in a tabular and machine-readable format. The top five NRs associated with the highest number of safety alerts from peer-reviewed journals, regulatory agency communications, congresses/conferences, clinical trial registries, and company communications were the Glucocorticoid Receptor (GR, 18,328), Androgen Receptor (AR, 18,219), Estrogen Receptor (ER, 12,028), Retinoic acid receptors (RAR, 10,450), and Pregnane X receptor (PXR, 8044). Toxicities associated with NR modulation include hepatotoxicity, cardiotoxicity, endocrine disruption, carcinogenicity, metabolic disorders, and neurotoxicity. These toxicities often arise from the dysregulation of receptors like Peroxisome proliferator-activated receptors (PPARα, PPARγ), the ER, PXR, AR, and GR. This dysregulation leads to various health issues, including liver enlargement, hepatocellular carcinoma, heart-related problems, hormonal imbalances, tumor growth, metabolic syndromes, and brain function impairment. Gene expression analysis using heatmaps for human and rat tissues complemented the functional modulation of NRs associated with the reported toxicities. Interestingly, certain NRs showed ubiquitous expression in tissues not previously linked to toxicities, suggesting the potential utilization of organ-specific NR interactions for therapeutic purposes.
Collapse
Affiliation(s)
- Mohan Rao
- Toxicology Department, Neurocrine Biosciences, Inc., San Diego, CA 92130, USA (C.S.)
| | - Eric McDuffie
- Toxicology Department, Neurocrine Biosciences, Inc., San Diego, CA 92130, USA (C.S.)
| | - Sanjay Srivastava
- Chemistry Department, Neurocrine Biosciences, Inc., San Diego, CA 92130, USA
| | - Warren Plaisted
- Biology Department, Neurocrine Biosciences, Inc., San Diego, CA 92130, USA
| | - Clifford Sachs
- Toxicology Department, Neurocrine Biosciences, Inc., San Diego, CA 92130, USA (C.S.)
| |
Collapse
|
12
|
Liu J, Gui Y, Rao J, Sun J, Wang G, Ren Q, Qu N, Niu B, Chen Z, Sheng X, Wang Y, Zheng M, Li X. In silico off-target profiling for enhanced drug safety assessment. Acta Pharm Sin B 2024; 14:2927-2941. [PMID: 39027254 PMCID: PMC11252485 DOI: 10.1016/j.apsb.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/21/2024] [Accepted: 02/29/2024] [Indexed: 07/20/2024] Open
Abstract
Ensuring drug safety in the early stages of drug development is crucial to avoid costly failures in subsequent phases. However, the economic burden associated with detecting drug off-targets and potential side effects through in vitro safety screening and animal testing is substantial. Drug off-target interactions, along with the adverse drug reactions they induce, are significant factors affecting drug safety. To assess the liability of candidate drugs, we developed an artificial intelligence model for the precise prediction of compound off-target interactions, leveraging multi-task graph neural networks. The outcomes of off-target predictions can serve as representations for compounds, enabling the differentiation of drugs under various ATC codes and the classification of compound toxicity. Furthermore, the predicted off-target profiles are employed in adverse drug reaction (ADR) enrichment analysis, facilitating the inference of potential ADRs for a drug. Using the withdrawn drug Pergolide as an example, we elucidate the mechanisms underlying ADRs at the target level, contributing to the exploration of the potential clinical relevance of newly predicted off-target interactions. Overall, our work facilitates the early assessment of compound safety/toxicity based on off-target identification, deduces potential ADRs of drugs, and ultimately promotes the secure development of drugs.
Collapse
Affiliation(s)
- Jin Liu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
| | - Yike Gui
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
- Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jingxin Rao
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingjing Sun
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Gang Wang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qun Ren
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
- Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ning Qu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Buying Niu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhiyi Chen
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, Hangzhou 330106, China
| | - Xia Sheng
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yitian Wang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mingyue Zheng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Nanjing University of Chinese Medicine, Nanjing 210023, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, Hangzhou 330106, China
| | - Xutong Li
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
13
|
Cichońska A, Ravikumar B, Rahman R. AI for targeted polypharmacology: The next frontier in drug discovery. Curr Opin Struct Biol 2024; 84:102771. [PMID: 38215530 DOI: 10.1016/j.sbi.2023.102771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/30/2023] [Accepted: 12/20/2023] [Indexed: 01/14/2024]
Abstract
In drug discovery, targeted polypharmacology, i.e., targeting multiple molecular targets with a single drug, is redefining therapeutic design to address complex diseases. Pre-selected pharmacological profiles, as exemplified in kinase drugs, promise enhanced efficacy and reduced toxicity. Historically, many of such drugs were discovered serendipitously, limiting predictability and efficacy, but currently artificial intelligence (AI) offers a transformative solution. Machine learning and deep learning techniques enable modeling protein structures, generating novel compounds, and decoding their polypharmacological effects, opening an avenue for more systematic and predictive multi-target drug design. This review explores the use of AI in identifying synergistic co-targets and delineating them from anti-targets that lead to adverse effects, and then discusses advances in AI-enabled docking, generative chemistry, and proteochemometric modeling of proteome-wide compound interactions, in the context of polypharmacology. We also provide insights into challenges ahead.
Collapse
|