1
|
Spanakis M, Tzamali E, Tzedakis G, Koumpouzi C, Pediaditis M, Tsatsakis A, Sakkalis V. Artificial Intelligence Models and Tools for the Assessment of Drug-Herb Interactions. Pharmaceuticals (Basel) 2025; 18:282. [PMID: 40143062 PMCID: PMC11944892 DOI: 10.3390/ph18030282] [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: 01/14/2025] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 03/28/2025] Open
Abstract
Artificial intelligence (AI) has emerged as a powerful tool in medical sciences that is revolutionizing various fields of drug research. AI algorithms can analyze large-scale biological data and identify molecular targets and pathways advancing pharmacological knowledge. An especially promising area is the assessment of drug interactions. The AI analysis of large datasets, such as drugs' chemical structure, pharmacological properties, molecular pathways, and known interaction patterns, can provide mechanistic insights and identify potential associations by integrating all this complex information and returning potential risks associated with these interactions. In this context, an area where AI may prove valuable is in the assessment of the underlying mechanisms of drug interactions with natural products (i.e., herbs) that are used as dietary supplements. These products pose a challenging problem since they are complex mixtures of constituents with diverse and limited information regarding their pharmacological properties, especially their pharmacokinetic data. As the use of herbal products and supplements continues to grow, it becomes increasingly important to understand the potential interactions between them and conventional drugs and the associated adverse drug reactions. This review will discuss AI approaches and how they can be exploited in providing valuable mechanistic insights regarding the prediction of interactions between drugs and herbs, and their potential exploitation in experimental validation or clinical utilization.
Collapse
Affiliation(s)
- Marios Spanakis
- Department of Toxicology and Forensic Sciences, School of Medicine, University of Crete, 71003 Heraklion, Greece;
- Computational Bio-Medicine Laboratory, Institute of Computer Science, Foundation for Research and Technology—Hellas, 70013 Heraklion, Greece; (E.T.); (G.T.); (C.K.); (M.P.); (V.S.)
| | - Eleftheria Tzamali
- Computational Bio-Medicine Laboratory, Institute of Computer Science, Foundation for Research and Technology—Hellas, 70013 Heraklion, Greece; (E.T.); (G.T.); (C.K.); (M.P.); (V.S.)
| | - Georgios Tzedakis
- Computational Bio-Medicine Laboratory, Institute of Computer Science, Foundation for Research and Technology—Hellas, 70013 Heraklion, Greece; (E.T.); (G.T.); (C.K.); (M.P.); (V.S.)
| | - Chryssalenia Koumpouzi
- Computational Bio-Medicine Laboratory, Institute of Computer Science, Foundation for Research and Technology—Hellas, 70013 Heraklion, Greece; (E.T.); (G.T.); (C.K.); (M.P.); (V.S.)
| | - Matthew Pediaditis
- Computational Bio-Medicine Laboratory, Institute of Computer Science, Foundation for Research and Technology—Hellas, 70013 Heraklion, Greece; (E.T.); (G.T.); (C.K.); (M.P.); (V.S.)
| | - Aristides Tsatsakis
- Department of Toxicology and Forensic Sciences, School of Medicine, University of Crete, 71003 Heraklion, Greece;
| | - Vangelis Sakkalis
- Computational Bio-Medicine Laboratory, Institute of Computer Science, Foundation for Research and Technology—Hellas, 70013 Heraklion, Greece; (E.T.); (G.T.); (C.K.); (M.P.); (V.S.)
| |
Collapse
|
2
|
Pelkonen O. Drug Metabolism - FromIn VitrotoIn Vivo, From Simple to Complex. Basic Clin Pharmacol Toxicol 2015; 117:147-55. [DOI: 10.1111/bcpt.12429] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 06/09/2015] [Indexed: 11/26/2022]
Affiliation(s)
- Olavi Pelkonen
- Centre of Biomedical Research; Department of Pharmacology and Toxicology; University of Oulu; Oulu Finland
| |
Collapse
|
3
|
Gundert-Remy U, Bernauer U, Blömeke B, Döring B, Fabian E, Goebel C, Hessel S, Jäckh C, Lampen A, Oesch F, Petzinger E, Völkel W, Roos PH. Extrahepatic metabolism at the body's internal–external interfaces. Drug Metab Rev 2014; 46:291-324. [DOI: 10.3109/03602532.2014.900565] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
4
|
Kapitulnik J. Drug transport and metabolism in the blood-brain barrier. Front Pharmacol 2011; 2:37. [PMID: 21811462 PMCID: PMC3139925 DOI: 10.3389/fphar.2011.00037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 07/05/2011] [Indexed: 11/23/2022] Open
Affiliation(s)
- Jaime Kapitulnik
- Department of Pharmacology, Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem Jerusalem, Israel
| |
Collapse
|
5
|
Adler S, Basketter D, Creton S, Pelkonen O, van Benthem J, Zuang V, Andersen KE, Angers-Loustau A, Aptula A, Bal-Price A, Benfenati E, Bernauer U, Bessems J, Bois FY, Boobis A, Brandon E, Bremer S, Broschard T, Casati S, Coecke S, Corvi R, Cronin M, Daston G, Dekant W, Felter S, Grignard E, Gundert-Remy U, Heinonen T, Kimber I, Kleinjans J, Komulainen H, Kreiling R, Kreysa J, Leite SB, Loizou G, Maxwell G, Mazzatorta P, Munn S, Pfuhler S, Phrakonkham P, Piersma A, Poth A, Prieto P, Repetto G, Rogiers V, Schoeters G, Schwarz M, Serafimova R, Tähti H, Testai E, van Delft J, van Loveren H, Vinken M, Worth A, Zaldivar JM. Alternative (non-animal) methods for cosmetics testing: current status and future prospects-2010. Arch Toxicol 2011; 85:367-485. [PMID: 21533817 DOI: 10.1007/s00204-011-0693-2] [Citation(s) in RCA: 358] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Accepted: 03/03/2011] [Indexed: 01/09/2023]
Abstract
The 7th amendment to the EU Cosmetics Directive prohibits to put animal-tested cosmetics on the market in Europe after 2013. In that context, the European Commission invited stakeholder bodies (industry, non-governmental organisations, EU Member States, and the Commission's Scientific Committee on Consumer Safety) to identify scientific experts in five toxicological areas, i.e. toxicokinetics, repeated dose toxicity, carcinogenicity, skin sensitisation, and reproductive toxicity for which the Directive foresees that the 2013 deadline could be further extended in case alternative and validated methods would not be available in time. The selected experts were asked to analyse the status and prospects of alternative methods and to provide a scientifically sound estimate of the time necessary to achieve full replacement of animal testing. In summary, the experts confirmed that it will take at least another 7-9 years for the replacement of the current in vivo animal tests used for the safety assessment of cosmetic ingredients for skin sensitisation. However, the experts were also of the opinion that alternative methods may be able to give hazard information, i.e. to differentiate between sensitisers and non-sensitisers, ahead of 2017. This would, however, not provide the complete picture of what is a safe exposure because the relative potency of a sensitiser would not be known. For toxicokinetics, the timeframe was 5-7 years to develop the models still lacking to predict lung absorption and renal/biliary excretion, and even longer to integrate the methods to fully replace the animal toxicokinetic models. For the systemic toxicological endpoints of repeated dose toxicity, carcinogenicity and reproductive toxicity, the time horizon for full replacement could not be estimated.
Collapse
Affiliation(s)
- Sarah Adler
- Centre for Documentation and Evaluation of Alternatives to Animal Experiments (ZEBET), Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Salminen KA, Meyer A, Jerabkova L, Korhonen LE, Rahnasto M, Juvonen RO, Imming P, Raunio H. Inhibition of human drug metabolizing cytochrome P450 enzymes by plant isoquinoline alkaloids. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2011; 18:533-538. [PMID: 20851588 DOI: 10.1016/j.phymed.2010.08.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Revised: 07/01/2010] [Accepted: 08/11/2010] [Indexed: 05/29/2023]
Abstract
The human cytochrome P450 (CYP) enzymes play a major role in the metabolism of endobiotics and numerous xenobiotics including drugs. Therefore it is the standard procedure to test new drug candidates for interactions with CYP enzymes during the preclinical development phase. The purpose of this study was to determine in vitro CYP inhibition potencies of a set of isoquinoline alkaloids to gain insight into interactions of novel chemical structures with CYP enzymes. These alkaloids (n=36) consist of compounds isolated from the Papaveraceae family (n=20), synthetic analogs (n=15), and one commercial compound. Their inhibitory activity was determined towards all principal human drug metabolizing CYP enzymes: 1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6 and 3A4. All alkaloids were assayed in vitro in a 96-well plate format using pro-fluorescent probe substrates and recombinant human CYP enzymes. Many of these alkaloids inhibited the CYP3A4 form, with 30/36 alkaloids inhibiting CYP3A4 with at least moderate potency (IC₅₀ < 10 μM) and 15/36 inhibiting CYP3A4 potently (IC₅₀ < 1 μM). Among them corydine, parfumine and 8-methyl-2,3,10,11-tetraethoxyberbine were potent and selective inhibitors for CYP3A4. CYP2D6 was inhibited with at least moderate potency by 26/34 alkaloids. CYP2C19 was inhibited by 15/36 alkaloids at least moderate potently, whereas CYP1A2, CYP2B6, CYP2C8, and CYP2C9 were inhibited to a lesser degree. CYP2A6 was not significantly inhibited by any of the alkaloids. The results provide initial structure-activity information about the interaction of isoquinoline alkaloids with major human xenobiotic-metabolizing CYP enzymes, and illustrate potential novel structures as CYP form-selective inhibitors.
Collapse
Affiliation(s)
- Kaisa A Salminen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Wittenburg LA, Gustafson DL. Optimizing preclinical study design in oncology research. Chem Biol Interact 2011; 190:73-8. [PMID: 21296059 DOI: 10.1016/j.cbi.2011.01.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Revised: 01/07/2011] [Accepted: 01/26/2011] [Indexed: 10/18/2022]
Abstract
The current drug development pathway in oncology research has led to a large attrition rate for new drugs, in part due to a general lack of appropriate preclinical studies that are capable of accurately predicting efficacy and/or toxicity in the target population. Because of an obvious need for novel therapeutics in many types of cancer, new compounds are being investigated in human Phase I and Phase II clinical trials before a complete understanding of their toxicity and efficacy profiles is obtained. In fact, for newer targeted molecular agents that are often cytostatic in nature, the conventional preclinical evaluation used for traditional cytotoxic chemotherapies utilizing primary tumor shrinkage as an endpoint may not be appropriate. By utilizing an integrated pharmacokinetic/pharmacodynamic approach, along with proper selection of a model system, the drug development process in oncology research may be improved leading to a better understanding of the determinants of efficacy and toxicity, and ultimately fewer drugs that fail once they reach human clinical trials.
Collapse
Affiliation(s)
- Luke A Wittenburg
- Flint Animal Cancer Center, Department of Clinical Sciences, Colorado State University, 300 West Drake Road, Fort Collins, CO 80523-1620, United States.
| | | |
Collapse
|
8
|
Kloft C, Poggesi I. Current and future directions of pharmacokinetic and pharmacokinetic-pharmacodynamic modelling and simulation: population approach group in Europe 19th annual meeting. Expert Opin Drug Metab Toxicol 2010; 6:1599-604. [PMID: 20969486 DOI: 10.1517/17425255.2010.529899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The Population Approach Group in Europe meeting is the annual meeting of a group of scientists interested in quantitative pharmacology approaches. This meeting provides insights on new modelling approaches and applications thereof in different therapeutic areas and in different phases of drug development, in which modelling and simulation is set 'at work'. These meeting highlights mainly focus on the contributions pertinent to this journal, such as physiology-based pharmacokinetic modelling, modelling of pharmacokinetic-pharmacodynamic relationships (including binary/categorical end points) in non-clinical studies and also include presentations of junior scientists in the annual Lewis Sheiner Student Session.
Collapse
Affiliation(s)
- Charlotte Kloft
- Institute of Pharmacy, Department of Clinical Pharmacy, Martin-Luther-Universitaet Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle, Germany
| | | |
Collapse
|