1
|
Zhai J, Man VH, Ji B, Cai L, Wang J. Comparison and summary of in silico prediction tools for CYP450-mediated drug metabolism. Drug Discov Today 2023; 28:103728. [PMID: 37517604 PMCID: PMC10543639 DOI: 10.1016/j.drudis.2023.103728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/30/2023] [Accepted: 07/25/2023] [Indexed: 08/01/2023]
Abstract
The cytochrome P450 (CYP450) enzyme system is responsible for the metabolism of more than two-thirds of xenobiotics. This review summarizes reports of a series of in silico tools for CYP450 enzyme-drug interaction predictions, including the prediction of sites of metabolism (SOM) of a drug and the identification of inhibitor/substrates for CYP subtypes. We also evaluated four prediction tools to identify CYP inhibitors utilizing 52 of the most frequently prescribed drugs. ADMET Predictor and CYPlebrity demonstrated the best performance. We hope that this review provides guidance for choosing appropriate enzyme prediction tools from a variety of in silico platforms to meet individual needs. Such predictions are useful for medicinal chemists to prioritize their designed compounds for further drug discovery.
Collapse
Affiliation(s)
- Jingchen Zhai
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Viet Hoang Man
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Beihong Ji
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Lianjin Cai
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Junmei Wang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
2
|
Chakravarti S. Computational Prediction of Metabolic α-Carbon Hydroxylation Potential of N-Nitrosamines: Overcoming Data Limitations for Carcinogenicity Assessment. Chem Res Toxicol 2023. [PMID: 37267457 DOI: 10.1021/acs.chemrestox.3c00083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Recent withdrawal of several drugs from the market due to elevated levels of N-nitrosamine impurities underscores the need for computational approaches to assess the carcinogenicity risk of nitrosamines. However, current approaches are limited because robust animal carcinogenicity data are only available for a few simple nitrosamines, which do not represent the structural diversity of the many possible nitrosamine drug substance related impurities (NDSRIs). In this paper, we present a novel method that uses data on CYP-mediated metabolic hydroxylation of CH2 groups in non-nitrosamine xenobiotics to identify structural features that may also help in predicting the likelihood of metabolic α-carbon hydroxylation in N-nitrosamines. Our approach offers a new avenue for tapping into potentially large experimental data sets on xenobiotic metabolism to improve risk assessment of nitrosamines. As α-carbon hydroxylation is the crucial rate-limiting step in nitrosamine metabolic activation, identifying and quantifying the influence of various structural features on this step can provide valuable insights into their carcinogenic potential. This is especially important considering the scarce information available on factors that affect NDSRI metabolic activation. We have identified hundreds of structural features and calculated their impact on hydroxylation, a significant advancement compared to the limited findings from the small nitrosamine carcinogenicity data set. While relying solely on α-carbon hydroxylation prediction is insufficient for forecasting carcinogenic potency, the identified features can help in the selection of relevant structural analogues in read across studies and assist experts who, after considering other factors such as the reactivity of the resulting electrophilic diazonium species, can establish the acceptable intake (AI) limits for nitrosamine impurities.
Collapse
Affiliation(s)
- Suman Chakravarti
- MultiCASE Inc., 23811 Chagrin Blvd, Suite 305, Beachwood, Ohio 44122, United States
| |
Collapse
|
3
|
Mitkov J, Kondeva-Burdina M, Georgieva M, Zlatkov A. Synthesis, in silico prediction of sites of metabolism and in-vitro hepatotoxicity evaluationofnew seriesN’-substituted 3-(1,3,7-trimethyl-xanthin-8-ylthio)propanehydrazides. PHARMACIA 2022. [DOI: 10.3897/pharmacia.69.e83535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
New series of 3-(1,3,7-trimethyl-xanthin-8-ylthio)propanehydrazide derivatives were designed and synthesized. The targed compounds were obtained in yields of 54 to 100% and their structures were elucidated by FTIR,1H NMR,13C NMR, MS and microanalyses.
The tested compounds were subjected to in silico prediction of sites of metabolism (SOMs). The predictions show thatthe main metabolic changes will be primarily related to oxidation of the sulfur atom in the side chain, carried out under the action of CYP2C19, as well as O-demethylation of compounds containing methoxy groups.The N-demethylation of the xanthine fragment was determined to be regulated by CYP1A2, CYP2C9, CYP2D6 and CYP3A4. Theperformed in vitro studies confirmed for two of the tested compounds to be low hepatotoxic, due to the presented prooxidant effects at subcellular level (isolated rat liver microsomes). These results highlight these molecules as promising hydrazide-hydrazone structures for the design of compounds with low hepatotoxicity.
Collapse
|
4
|
Muller C, Rabal O, Diaz Gonzalez C. Artificial Intelligence, Machine Learning, and Deep Learning in Real-Life Drug Design Cases. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2021; 2390:383-407. [PMID: 34731478 DOI: 10.1007/978-1-0716-1787-8_16] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The discovery and development of drugs is a long and expensive process with a high attrition rate. Computational drug discovery contributes to ligand discovery and optimization, by using models that describe the properties of ligands and their interactions with biological targets. In recent years, artificial intelligence (AI) has made remarkable modeling progress, driven by new algorithms and by the increase in computing power and storage capacities, which allow the processing of large amounts of data in a short time. This review provides the current state of the art of AI methods applied to drug discovery, with a focus on structure- and ligand-based virtual screening, library design and high-throughput analysis, drug repurposing and drug sensitivity, de novo design, chemical reactions and synthetic accessibility, ADMET, and quantum mechanics.
Collapse
Affiliation(s)
- Christophe Muller
- Evotec (France) SAS, Computational Drug Discovery, Integrated Drug Discovery, Toulouse, France
| | - Obdulia Rabal
- Evotec (France) SAS, Computational Drug Discovery, Integrated Drug Discovery, Toulouse, France
| | | |
Collapse
|
5
|
Raju B, Choudhary S, Narendra G, Verma H, Silakari O. Molecular modeling approaches to address drug-metabolizing enzymes (DMEs) mediated chemoresistance: a review. Drug Metab Rev 2021; 53:45-75. [PMID: 33535824 DOI: 10.1080/03602532.2021.1874406] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Resistance against clinically approved anticancer drugs is the main roadblock in cancer treatment. Drug metabolizing enzymes (DMEs) that are capable of metabolizing a variety of xenobiotic get overexpressed in malignant cells, therefore, catalyzing drug inactivation. As evident from the literature reports, the levels of DMEs increase in cancer cells that ultimately lead to drug inactivation followed by drug resistance. To puzzle out this issue, several strategies inclusive of analog designing, prodrug designing, and inhibitor designing have been forged. On that front, the implementation of computational tools can be considered a fascinating approach to address the problem of chemoresistance. Various research groups have adopted different molecular modeling tools for the investigation of DMEs mediated toxicity problems. However, the utilization of these in-silico tools in maneuvering the DME mediated chemoresistance is least considered and yet to be explored. These tools can be employed in the designing of such chemotherapeutic agents that are devoid of the resistance problem. The current review canvasses various molecular modeling approaches that can be implemented to address this issue. Special focus was laid on the development of specific inhibitors of DMEs. Additionally, the strategies to bypass the DMEs mediated drug metabolism were also contemplated in this report that includes analogs and pro-drugs designing. Different strategies discussed in the review will be beneficial in designing novel chemotherapeutic agents that depreciate the resistance problem.
Collapse
Affiliation(s)
- Baddipadige Raju
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Shalki Choudhary
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Gera Narendra
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Himanshu Verma
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Om Silakari
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| |
Collapse
|
6
|
Filimonov DA, Rudik AV, Dmitriev AV, Lagunin AA, Poroikov VV. Computer Assessment of the Xenobiotic Metabolites Formation’s Probability in the Human Body. Biophysics (Nagoya-shi) 2020. [DOI: 10.1134/s0006350920060044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
7
|
Poroikov VV. Computer-Aided Drug Design: from Discovery of Novel Pharmaceutical Agents to Systems Pharmacology. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES B: BIOMEDICAL CHEMISTRY 2020. [DOI: 10.1134/s1990750820030117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
8
|
Flynn NR, Dang NL, Ward MD, Swamidass SJ. XenoNet: Inference and Likelihood of Intermediate Metabolite Formation. J Chem Inf Model 2020; 60:3431-3449. [PMID: 32525671 PMCID: PMC8716322 DOI: 10.1021/acs.jcim.0c00361] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Drug metabolism is a common cause of adverse drug reactions. Drug molecules can be metabolized into reactive metabolites, which can conjugate to biomolecules, like protein and DNA, in a process termed bioactivation. To mitigate adverse reactions caused by bioactivation, both experimental and computational screening assays are utilized. Experimental assays for assessing the formation of reactive metabolites are low throughput and expensive to perform, so they are often reserved until later stages of the drug development pipeline when the drug candidate pools are already significantly narrowed. In contrast, computational methods are high throughput and cheap to perform to screen thousands to millions of compounds for potentially toxic molecules during the early stages of the drug development pipeline. Commonly used computational methods focus on detecting and structurally characterizing reactive metabolite-biomolecule adducts or predicting sites on a drug molecule that are liable to form reactive metabolites. However, such methods are often only concerned with the structure of the initial drug molecule or of the adduct formed when a biomolecule conjugates to a reactive metabolite. Thus, these methods are likely to miss intermediate metabolites that may lead to subsequent reactive metabolite formation. To address these shortcomings, we create XenoNet, a metabolic network predictor, that can take a pair of a substrate and a target product as input and (1) enumerate pathways, or sequences of intermediate metabolite structures, between the pair, and (2) compute the likelihood of those pathways and intermediate metabolites. We validate XenoNet on a large, chemically diverse data set of 17 054 metabolic networks built from a literature-derived reaction database. Each metabolic network has a defined substrate molecule that has been experimentally observed to undergo metabolism into a defined product metabolite. XenoNet can predict experimentally observed pathways and intermediate metabolites linking the input substrate and product pair with a recall of 88 and 46%, respectively. Using likelihood scoring, XenoNet also achieves a top-one pathway and intermediate metabolite accuracy of 93.6 and 51.9%, respectively. We further validate XenoNet against prior methods for metabolite prediction. XenoNet significantly outperforms all prior methods across multiple metrics. XenoNet is available at https://swami.wustl.edu/xenonet.
Collapse
Affiliation(s)
- Noah R Flynn
- Department of Pathology and Immunology, Washington University School of Medicine, Campus Box 8118, 660 S. Euclid Ave., St. Louis, Missouri 63110, United States
| | - Na Le Dang
- Department of Pathology and Immunology, Washington University School of Medicine, Campus Box 8118, 660 S. Euclid Ave., St. Louis, Missouri 63110, United States
| | - Michael D Ward
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S Euclid Ave, St. Louis, Missouri 63110, United States
| | - S Joshua Swamidass
- Department of Pathology and Immunology, Washington University School of Medicine, Campus Box 8118, 660 S. Euclid Ave., St. Louis, Missouri 63110, United States
| |
Collapse
|
9
|
Poroikov VV. [Computer-aided drug design: from discovery of novel pharmaceutical agents to systems pharmacology]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2020; 66:30-41. [PMID: 32116224 DOI: 10.18097/pbmc20206601030] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
New drug discovery is based on the analysis of public information about the mechanisms of the disease, molecular targets, and ligands, which interaction with the target could lead to the normalization of the pathological process. The available data on diseases, drugs, pharmacological effects, molecular targets, and drug-like substances, taking into account the combinatorics of the associative relations between them, correspond to the Big Data. To analyze such data, the application of computer-aided drug design methods is necessary. An overview of the studies in this area performed by the Laboratory for Structure-Function Based Drug Design of IBMC is presented. We have developed the approaches to identifying promising pharmacological targets, predicting several thousand types of biological activity based on the structural formula of the compound, analyzing protein-ligand interactions based on assessing local similarity of amino acid sequences, identifying likely molecular mechanisms of side effects of drugs, calculating the integral toxicity of drugs taking into account their metabolism, have been developed in the human body, predicting sustainable and sensitive options strains and evaluating the effectiveness of combinations of antiretroviral drugs in patients, taking into account the molecular genetic characteristics of the clinical isolates of HIV-1. Our computer programs are implemented as the web-services freely available on the Internet, which are used by thousands of researchers from many countries of the world to select the most promising substances for the synthesis and determine the priority areas for experimental testing of their biological activity.
Collapse
Affiliation(s)
- V V Poroikov
- Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
10
|
Dang NL, Matlock MK, Hughes TB, Swamidass SJ. The Metabolic Rainbow: Deep Learning Phase I Metabolism in Five Colors. J Chem Inf Model 2020; 60:1146-1164. [DOI: 10.1021/acs.jcim.9b00836] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Na Le Dang
- Department of Pathology and Immunology, Washington University School of Medicine, Campus Box 8118, 660 S. Euclid Ave., St. Louis, Missouri 63110, United States
| | - Matthew K. Matlock
- Department of Pathology and Immunology, Washington University School of Medicine, Campus Box 8118, 660 S. Euclid Ave., St. Louis, Missouri 63110, United States
| | - Tyler B. Hughes
- Department of Pathology and Immunology, Washington University School of Medicine, Campus Box 8118, 660 S. Euclid Ave., St. Louis, Missouri 63110, United States
| | - S. Joshua Swamidass
- Department of Pathology and Immunology, Washington University School of Medicine, Campus Box 8118, 660 S. Euclid Ave., St. Louis, Missouri 63110, United States
| |
Collapse
|
11
|
Rudik A, Bezhentsev V, Dmitriev A, Lagunin A, Filimonov D, Poroikov V. Metatox - Web application for generation of metabolic pathways and toxicity estimation. J Bioinform Comput Biol 2020; 17:1940001. [PMID: 30866738 DOI: 10.1142/s0219720019400018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Xenobiotics biotransformation in humans is a process of the chemical modifications, which may lead to the formation of toxic metabolites. The prediction of such metabolites is very important for drug development and ecotoxicology studies. We created the web-application MetaTox ( http://way2drug.com/mg ) for the generation of xenobiotics metabolic pathways in the human organism. For each generated metabolite, the estimations of the acute toxicity (based on GUSAR software prediction), organ-specific carcinogenicity and adverse effects (based on PASS software prediction) are performed. Generation of metabolites by MetaTox is based on the fragments datasets, which describe transformations of substrates structures to a metabolites structure. We added three new classes of biotransformation reactions: Dehydrogenation, Glutathionation, and Hydrolysis, and now metabolite generation for 15 most frequent classes of xenobiotic's biotransformation reactions are available. MetaTox calculates the probability of formation of generated metabolite - it is the integrated assessment of the biotransformation reactions probabilities and their sites using the algorithm of PASS ( http://way2drug.com/passonline ). The prediction accuracy estimated by the leave-one-out cross-validation (LOO-CV) procedure calculated separately for the probabilities of biotransformation reactions and their sites is about 0.9 on the average for all reactions.
Collapse
Affiliation(s)
- Anastasiya Rudik
- * Department of Bioinformatics, Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, Moscow 119121, Russia
| | - Vladislav Bezhentsev
- * Department of Bioinformatics, Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, Moscow 119121, Russia
| | - Alexander Dmitriev
- * Department of Bioinformatics, Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, Moscow 119121, Russia
| | - Alexey Lagunin
- * Department of Bioinformatics, Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, Moscow 119121, Russia.,† Medico-Biological Faculty, Pirogov Russian National Research Medical University, 1 Ostrovitianov Street, Moscow 117997, Russia
| | - Dmitry Filimonov
- * Department of Bioinformatics, Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, Moscow 119121, Russia
| | - Vladimir Poroikov
- * Department of Bioinformatics, Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, Moscow 119121, Russia
| |
Collapse
|
12
|
Computer-aided prediction of biological activity spectra for organic compounds: the possibilities and limitations. Russ Chem Bull 2020. [DOI: 10.1007/s11172-019-2683-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
13
|
de Sá Hyacienth BM, Tavares Picanço KR, Sánchez-Ortiz BL, Barros Silva L, Matias Pereira AC, Machado Góes LD, Sousa Borges R, Cardoso Ataíde R, dos Santos CBR, de Oliveira Carvalho H, Gonzalez Anduaga GM, Navarrete A, Tavares Carvalho JC. Hydroethanolic extract from Endopleura uchi (Huber) Cuatrecasas and its marker bergenin: Toxicological and pharmacokinetic studies in silico and in vivo on zebrafish. Toxicol Rep 2020; 7:217-232. [PMID: 32042599 PMCID: PMC6997909 DOI: 10.1016/j.toxrep.2020.01.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 01/24/2020] [Accepted: 01/26/2020] [Indexed: 02/07/2023] Open
Abstract
E. uchi stem bark hydroethanolic extract in zebrafish. Evaluating the in silico pharmacokinetic and toxicological parameters. Behavioral, biochemical and histopathological changes was dose dependent. In silico bergenin and its metabolites showed high intestinal absorption. Bergenin inhibited CYP2C9, CYP3A4 and CYP2C19.
Endopleura uchi, is used for the treatment of inflammatory disease and related to the female reproductive tract. The aim of this study was to evaluate the acute toxicity of the Endopleura uchi stem bark hydroethanolic extract (EEu) in zebrafish, emphasizing the histopathological and biochemical parameters, as well as evaluating the in silico pharmacokinetic and toxicological parameters of the phytochemical/pharmacological marker, bergenin, as their metabolites. The animals were orally treated with EEu at a single dose of 75 mg/kg, 500 mg/kg, 1000 mg/kg and 3000 mg/kg. the oral LD50 of the EEu higher to the dose of 3000 mg/kg. Behavioral, biochemical and histopathological changes were dose dependent. In silico pharmacokinetic predictions for bergenin and its metabolites showed moderate absorption in high human intestinal absorption (HIA) and Caco-2 models, reduced plasma protein binding, by low brain tissue binding and no P-glycoprotein (P-Gp) inhibition. Their metabolism is defined by the CYP450 enzyme, in addition to bergenin inhibition of CYP2C9, CYP3A4 and CYP2C19. In the bergenin and its metabolites in silico toxicity test it have been shown to cause carcinogenicity and a greater involvement of the bergenin with the CYP enzymes in the I and II hepatic and renal metabolism’s phases was observed. It is possible to suggest that the histopathological damages are involved with the interaction of this major compound and its metabolites at the level of the cellular-biochemical mechanisms which involve the absorption, metabolization and excretion of these possible prodrug and drug.
Collapse
Key Words
- ALT, Alanine aminotransferase
- AST, Aspartate aminotransferase
- BBB, Brain-blood partition coefficient (C.brain/C.blood)
- Bergenin
- Biotrasformation
- EEu, Endopleura uchi stem bark hydroethanolic extract
- Endopleura uchi
- HAI, Index of Histopathological Changes
- HBA, Hydrogen bonding acceptors
- HBD, Hydrogen bonding donors
- HIA, Human intestinal absorption
- Hepatoxity
- IAN, Regional Herbarium of the Eastern Amazonian Embrapa
- MM, Molecular mass
- Nephrotoxity
- P-Gp, P-glycoprotein
- PPB, Plasma protein binding
- Toxicology
- hERG, ether-a-go-related human gene
Collapse
Affiliation(s)
- Beatriz Martins de Sá Hyacienth
- Laboratory of Pharmaceutical Research, Department of Biological Sciences and Health, Federal University of Amapá, Juscelino Kubitschek Street, Marco Zero Campus, Zip Code 68903-419, Macapá, AP, Brazil
- Postgraduate Program in Biodiversity and Biotechnology of the Legal Amazon of the BIONORTE Network, Department of Biological Sciences and Health, Federal University of Amapá, Macapá, AP, Brazil
| | - Karyny Roberta Tavares Picanço
- Laboratory of Pharmaceutical Research, Department of Biological Sciences and Health, Federal University of Amapá, Juscelino Kubitschek Street, Marco Zero Campus, Zip Code 68903-419, Macapá, AP, Brazil
| | - Brenda Lorena Sánchez-Ortiz
- Laboratory of Pharmaceutical Research, Department of Biological Sciences and Health, Federal University of Amapá, Juscelino Kubitschek Street, Marco Zero Campus, Zip Code 68903-419, Macapá, AP, Brazil
- Laboratory of Natural Product Pharmacology, Department of Pharmacy, Faculty of Chemistry, National Autonomous University of Mexico, University City, Coyoacán, Zip Code 04510 Mexico City, Mexico
| | - Luciane Barros Silva
- Federal University of Amapá, Laboratory of Modeling and Computational Chemistry, Department of Biological Sciences and Health, Juscelino Kubitschek Street, Marco Zero Campus, Zip Code 68903-419, Macapá, AP, Brazil
| | - Arlindo César Matias Pereira
- Laboratory of Pharmaceutical Research, Department of Biological Sciences and Health, Federal University of Amapá, Juscelino Kubitschek Street, Marco Zero Campus, Zip Code 68903-419, Macapá, AP, Brazil
| | - Larissa Daniele Machado Góes
- Laboratory of Pharmaceutical Research, Department of Biological Sciences and Health, Federal University of Amapá, Juscelino Kubitschek Street, Marco Zero Campus, Zip Code 68903-419, Macapá, AP, Brazil
| | - Raphaelle Sousa Borges
- Laboratory of Pharmaceutical Research, Department of Biological Sciences and Health, Federal University of Amapá, Juscelino Kubitschek Street, Marco Zero Campus, Zip Code 68903-419, Macapá, AP, Brazil
| | - Rodrigo Cardoso Ataíde
- Laboratory of Pharmaceutical Research, Department of Biological Sciences and Health, Federal University of Amapá, Juscelino Kubitschek Street, Marco Zero Campus, Zip Code 68903-419, Macapá, AP, Brazil
| | - Cleydson Breno Rodrigues dos Santos
- Laboratory of Pharmaceutical Research, Department of Biological Sciences and Health, Federal University of Amapá, Juscelino Kubitschek Street, Marco Zero Campus, Zip Code 68903-419, Macapá, AP, Brazil
- Federal University of Amapá, Laboratory of Modeling and Computational Chemistry, Department of Biological Sciences and Health, Juscelino Kubitschek Street, Marco Zero Campus, Zip Code 68903-419, Macapá, AP, Brazil
| | - Helison de Oliveira Carvalho
- Laboratory of Pharmaceutical Research, Department of Biological Sciences and Health, Federal University of Amapá, Juscelino Kubitschek Street, Marco Zero Campus, Zip Code 68903-419, Macapá, AP, Brazil
| | - Gloria Melisa Gonzalez Anduaga
- Laboratory of Natural Product Pharmacology, Department of Pharmacy, Faculty of Chemistry, National Autonomous University of Mexico, University City, Coyoacán, Zip Code 04510 Mexico City, Mexico
| | - Andrés Navarrete
- Laboratory of Natural Product Pharmacology, Department of Pharmacy, Faculty of Chemistry, National Autonomous University of Mexico, University City, Coyoacán, Zip Code 04510 Mexico City, Mexico
| | - José Carlos Tavares Carvalho
- Laboratory of Pharmaceutical Research, Department of Biological Sciences and Health, Federal University of Amapá, Juscelino Kubitschek Street, Marco Zero Campus, Zip Code 68903-419, Macapá, AP, Brazil
- Postgraduate Program in Biodiversity and Biotechnology of the Legal Amazon of the BIONORTE Network, Department of Biological Sciences and Health, Federal University of Amapá, Macapá, AP, Brazil
- Corresponding author.
| |
Collapse
|
14
|
Synthesis and biological evaluation of new multi-target 3-(1H-indol-3-yl)pyrrolidine-2,5-dione derivatives with potential antidepressant effect. Eur J Med Chem 2019; 183:111736. [DOI: 10.1016/j.ejmech.2019.111736] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 12/31/2022]
|
15
|
Bruno A, Costantino G, Sartori L, Radi M. The In Silico Drug Discovery Toolbox: Applications in Lead Discovery and Optimization. Curr Med Chem 2019; 26:3838-3873. [PMID: 29110597 DOI: 10.2174/0929867324666171107101035] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/27/2017] [Accepted: 09/28/2017] [Indexed: 01/04/2023]
Abstract
BACKGROUND Discovery and development of a new drug is a long lasting and expensive journey that takes around 20 years from starting idea to approval and marketing of new medication. Despite R&D expenditures have been constantly increasing in the last few years, the number of new drugs introduced into market has been steadily declining. This is mainly due to preclinical and clinical safety issues, which still represent about 40% of drug discontinuation. To cope with this issue, a number of in silico techniques are currently being used for an early stage evaluation/prediction of potential safety issues, allowing to increase the drug-discovery success rate and reduce costs associated with the development of a new drug. METHODS In the present review, we will analyse the early steps of the drug-discovery pipeline, describing the sequence of steps from disease selection to lead optimization and focusing on the most common in silico tools used to assess attrition risks and build a mitigation plan. RESULTS A comprehensive list of widely used in silico tools, databases, and public initiatives that can be effectively implemented and used in the drug discovery pipeline has been provided. A few examples of how these tools can be problem-solving and how they may increase the success rate of a drug discovery and development program have been also provided. Finally, selected examples where the application of in silico tools had effectively contributed to the development of marketed drugs or clinical candidates will be given. CONCLUSION The in silico toolbox finds great application in every step of early drug discovery: (i) target identification and validation; (ii) hit identification; (iii) hit-to-lead; and (iv) lead optimization. Each of these steps has been described in details, providing a useful overview on the role played by in silico tools in the decision-making process to speed-up the discovery of new drugs.
Collapse
Affiliation(s)
- Agostino Bruno
- Experimental Therapeutics Unit, IFOM - The FIRC Institute for Molecular Oncology Foundation, Via Adamello 16 - 20139 Milano, Italy
| | - Gabriele Costantino
- Dipartimento di Scienze degli Alimenti e del Farmaco, Universita degli Studi di Parma, Viale delle Scienze, 27/A, 43124 Parma, Italy
| | - Luca Sartori
- Experimental Therapeutics Unit, IFOM - The FIRC Institute for Molecular Oncology Foundation, Via Adamello 16 - 20139 Milano, Italy
| | - Marco Radi
- Dipartimento di Scienze degli Alimenti e del Farmaco, Universita degli Studi di Parma, Viale delle Scienze, 27/A, 43124 Parma, Italy
| |
Collapse
|
16
|
Tyzack JD, Kirchmair J. Computational methods and tools to predict cytochrome P450 metabolism for drug discovery. Chem Biol Drug Des 2019; 93:377-386. [PMID: 30471192 PMCID: PMC6590657 DOI: 10.1111/cbdd.13445] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 11/05/2018] [Accepted: 11/11/2018] [Indexed: 01/08/2023]
Abstract
In this review, we present important, recent developments in the computational prediction of cytochrome P450 (CYP) metabolism in the context of drug discovery. We discuss in silico models for the various aspects of CYP metabolism prediction, including CYP substrate and inhibitor predictors, site of metabolism predictors (i.e., metabolically labile sites within potential substrates) and metabolite structure predictors. We summarize the different approaches taken by these models, such as rule‐based methods, machine learning, data mining, quantum chemical methods, molecular interaction fields, and docking. We highlight the scope and limitations of each method and discuss future implications for the field of metabolism prediction in drug discovery.
Collapse
Affiliation(s)
| | - Johannes Kirchmair
- Department of Chemistry, University of Bergen, Bergen, Norway.,Computational Biology Unit (CBU), University of Bergen, Bergen, Norway.,Center for Bioinformatics, Universität Hamburg, Hamburg, Germany
| |
Collapse
|
17
|
Ivashchenko DV, Rudik AV, Poloznikov AA, Nikulin SV, Smirnov VV, Tonevitsky AG, Bryun EA, Sychev DA. Which cytochrome P450 metabolizes phenazepam? Step by step in silico, in vitro, and in vivo studies. Drug Metab Pers Ther 2018; 33:65-73. [PMID: 29727298 DOI: 10.1515/dmpt-2017-0036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 02/03/2018] [Indexed: 01/17/2023]
Abstract
BACKGROUND Phenazepam (bromdihydrochlorphenylbenzodiazepine) is the original Russian benzodiazepine tranquilizer belonging to 1,4-benzodiazepines. There is still limited knowledge about phenazepam's metabolic liver pathways and other pharmacokinetic features. METHODS To determine phenazepam's metabolic pathways, the study was divided into three stages: in silico modeling, in vitro experiment (cell culture study), and in vivo confirmation. In silico modeling was performed on the specialized software PASS and GUSAR to evaluate phenazepam molecule affinity to different cytochromes. The in vitro study was performed using a hepatocytes' cell culture, cultivated in a microbioreactor to produce cytochrome P450 isoenzymes. The culture medium contained specific cytochrome P450 isoforms inhibitors and substrates (for CYP2C9, CYP3A4, CYP2C19, and CYP2B6) to determine the cytochrome that was responsible for phenazepam's metabolism. We also measured CYP3A activity using the 6-betahydroxycortisol/cortisol ratio in patients. RESULTS According to in silico and in vitro analysis results, the most probable metabolizer of phenazepam is CYP3A4. By the in vivo study results, CYP3A activity decreased sufficiently (from 3.8 [95% CI: 2.94-4.65] to 2.79 [95% CI: 2.02-3.55], p=0.017) between the start and finish of treatment in patients who were prescribed just phenazepam. CONCLUSIONS Experimental in silico and in vivo studies confirmed that the original Russian benzodiazepine phenazepam was the substrate of CYP3A4 isoenzyme.
Collapse
Affiliation(s)
- Dmitriy V Ivashchenko
- Russian Medical Academy of Continuous Professional Education, Moscow, Russian Federation
| | - Anastasia V Rudik
- Institute of Biomedical Chemistry, 10 bldg., Moscow, Russian Federation
| | - Andrey A Poloznikov
- Moscow Institute of Physics and Technology, 9 Institutsky lane, 141700, Dolgoprudny, Russian Federation;National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Koroleva, 4, 249036 Obninsk, Russian Federation;SRC "Bioclinicum", Moscow, Russian Federation
| | - Sergey V Nikulin
- Moscow Institute of Physics and Technology, 9 Institutsky lane, 141700, Dolgoprudny, Russian Federation;National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Koroleva, 4, 249036 Obninsk, Russian Federation;SRC "Bioclinicum", Moscow, Russian Federation
| | - Valeriy V Smirnov
- National Research Center - Institute of Immunology Federal Medical-Biological Agency of Russia, Moscow, Russian Federation
- I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Alexander G Tonevitsky
- Moscow Institute of Physics and Technology, 9 Institutsky lane, 141700, Dolgoprudny, Russian Federation;National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Koroleva, 4, 249036 Obninsk, Russian Federation;SRC "Bioclinicum", Moscow, Russian Federation
| | - Eugeniy A Bryun
- Russian Medical Academy of Continuous Professional Education, Moscow, Russian Federation
- Moscow Research Practical Center of Narcology, Moscow, Russian Federation
| | - Dmitriy A Sychev
- Russian Medical Academy of Continuous Professional Education, Moscow, Russian Federation
| |
Collapse
|
18
|
Fu X, He S, Du L, Lv Z, Zhang Y, Zhang Q, Wang Y. Using chemical bond-based method to predict site of metabolism for five biotransformations mediated by CYP 3A4, 2D6, and 2C9. Biochem Pharmacol 2018; 152:302-314. [PMID: 29588194 DOI: 10.1016/j.bcp.2018.03.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 03/22/2018] [Indexed: 11/29/2022]
Abstract
Although it has been proposed for decades to predict site of metabolism (SOM) by in silico methods, identifying SOM correctly remains an unsolved fundamental problem and is an active area of research. In our prior works, we proposed a chemical bond-based approach to construction of SOM prediction models by integrating chemical bond descriptors and drug-metabolizing enzymes data. Although it has been evaluated with both 10-fold cross-validation and independent validation, we believe comparisons between this method and prior methods using publicly accessible external datasets are indispensable and more desirable. In the current study, based on chemical bond-based method, metabolism data released by Sheridan et al. and Zaretzki et al. was utilized to establish metabolite prediction models for CYP450 3A4, 2D6, and 2C9. Five major reaction types were involved, including Aliphatic C-hydroxylation, Aromatic C-hydroxylation, N-dealkylation, O-dealkylation, and S-Oxidation. Consequently, all our five models showed impressive performance on predicting SOMs, with accuracy and area under curve exceeded 0.940 and 0.953, respectively. Compared to prior works, our models were better than SOMP both in "SOM-scale" and "molecule-scale". In conclusion, comparisons between chemical-bond based method and prior works were conducted for the first time, which demonstrated that chemical-bond based method is better than or at least comparable to prior works.
Collapse
Affiliation(s)
- XuYan Fu
- Key Laboratory of Traditional Chinese Medicine Information Engineer of State Administration of Traditional Chinese Medicine, School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China
| | - ShuaiBing He
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine from Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Li Du
- Key Laboratory of Traditional Chinese Medicine Information Engineer of State Administration of Traditional Chinese Medicine, School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China
| | - ZhaoLei Lv
- Key Laboratory of Traditional Chinese Medicine Information Engineer of State Administration of Traditional Chinese Medicine, School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Yi Zhang
- Key Laboratory of Traditional Chinese Medicine Information Engineer of State Administration of Traditional Chinese Medicine, School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Qian Zhang
- Key Laboratory of Traditional Chinese Medicine Information Engineer of State Administration of Traditional Chinese Medicine, School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Yun Wang
- Key Laboratory of Traditional Chinese Medicine Information Engineer of State Administration of Traditional Chinese Medicine, School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China.
| |
Collapse
|
19
|
Druzhilovskiy DS, Rudik AV, Filimonov DA, Gloriozova TA, Lagunin AA, Dmitriev AV, Pogodin PV, Dubovskaya VI, Ivanov SM, Tarasova OA, Bezhentsev VM, Murtazalieva KA, Semin MI, Maiorov IS, Gaur AS, Sastry GN, Poroikov VV. Computational platform Way2Drug: from the prediction of biological activity to drug repurposing. Russ Chem Bull 2018. [DOI: 10.1007/s11172-017-1954-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
20
|
Madzhidov TI, Khakimova AA, Nugmanov RI, Muller C, Marcou G, Varnek A. Prediction of Aromatic Hydroxylation Sites for Human CYP1A2 Substrates Using Condensed Graph of Reactions. BIONANOSCIENCE 2018. [DOI: 10.1007/s12668-017-0499-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
21
|
Abstract
Various methods of machine learning, supervised and unsupervised, linear and nonlinear, classification and regression, in combination with various types of molecular descriptors, both "handcrafted" and "data-driven," are considered in the context of their use in computational toxicology. The use of multiple linear regression, variants of naïve Bayes classifier, k-nearest neighbors, support vector machine, decision trees, ensemble learning, random forest, several types of neural networks, and deep learning is the focus of attention of this review. The role of fragment descriptors, graph mining, and graph kernels is highlighted. The application of unsupervised methods, such as Kohonen's self-organizing maps and related approaches, which allow for combining predictions with data analysis and visualization, is also considered. The necessity of applying a wide range of machine learning methods in computational toxicology is underlined.
Collapse
Affiliation(s)
- Igor I Baskin
- Faculty of Physics, M.V. Lomonosov Moscow State University, Moscow, Russian Federation.
- Butlerov Institute of Chemistry, Kazan Federal University, Kazan, Russian Federation.
| |
Collapse
|
22
|
Filimonov D, Druzhilovskiy D, Lagunin A, Gloriozova T, Rudik A, Dmitriev A, Pogodin P, Poroikov V. Computer-aided prediction of biological activity spectra for chemical compounds: opportunities and limitation. ACTA ACUST UNITED AC 2018. [DOI: 10.18097/bmcrm00004] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
An essential characteristic of chemical compounds is their biological activity since its presence can become the basis for the use of the substance for therapeutic purposes, or, on the contrary, limit the possibilities of its practical application due to the manifestation of side action and toxic effects. Computer assessment of the biological activity spectra makes it possible to determine the most promising directions for the study of the pharmacological action of particular substances, and to filter out potentially dangerous molecules at the early stages of research. For more than 25 years, we have been developing and improving the computer program PASS (Prediction of Activity Spectra for Substances), designed to predict the biological activity spectrum of substance based on the structural formula of its molecules. The prediction is carried out by the analysis of structure-activity relationships for the training set, which currently contains information on structures and known biological activities for more than one million molecules. The structure of the organic compound is represented in PASS using Multilevel Neighborhoods of Atoms descriptors; the activity prediction for new compounds is performed by the naive Bayes classifier and the structure-activity relationships determined by the analysis of the training set. We have created and improved both local versions of the PASS program and freely available web resources based on PASS (http://www.way2drug.com). They predict several thousand biological activities (pharmacological effects, molecular mechanisms of action, specific toxicity and adverse effects, interaction with the unwanted targets, metabolism and action on molecular transport), cytotoxicity for tumor and non-tumor cell lines, carcinogenicity, induced changes of gene expression profiles, metabolic sites of the major enzymes of the first and second phases of xenobiotics biotransformation, and belonging to substrates and/or metabolites of metabolic enzymes. The web resource Way2Drug is used by over 18,000 researchers from more than 90 countries around the world, which allowed them to obtain over 600,000 predictions and publish about 500 papers describing the obtained results. The analysis of the published works shows that in some cases the interpretation of the prediction results presented by the authors of these publications requires an adjustment. In this work, we provide the theoretical basis and consider, on particular examples, the opportunities and limitations of computer-aided prediction of biological activity spectra.
Collapse
Affiliation(s)
| | | | - A.A. Lagunin
- Institute of Biomedical Chemistry; Pirogov Russian National Research Medical University, Moscow, Russia
| | | | - A.V. Rudik
- Institute of Biomedical Chemistry, Moscow, Russia
| | | | - P.V. Pogodin
- Institute of Biomedical Chemistry, Moscow, Russia
| | | |
Collapse
|
23
|
Tarasova O, Rudik A, Dmitriev A, Lagunin A, Filimonov D, Poroikov V. QNA-Based Prediction of Sites of Metabolism. Molecules 2017; 22:molecules22122123. [PMID: 29194399 PMCID: PMC6149875 DOI: 10.3390/molecules22122123] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 11/28/2017] [Indexed: 01/25/2023] Open
Abstract
Metabolism of xenobiotics (Greek xenos: exogenous substances) plays an essential role in the prediction of biological activity and testing for the subsequent research and development of new drug candidates. Integration of various methods and techniques using different computational and experimental approaches is one of the keys to a successful metabolism prediction. While multiple structure-based and ligand-based approaches to metabolism prediction exist, the most important problem arises at the first stage of metabolism prediction: detection of the sites of metabolism (SOMs). In this paper, we describe the application of Quantitative Neighborhoods of Atoms (QNA) descriptors for prediction of the SOMs using potential function method, as well as several different machine learning techniques: naïve Bayes, random forest classifier, multilayer perceptron with back propagation and convolutional neural networks, and deep neural networks.
Collapse
Affiliation(s)
- Olga Tarasova
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia.
| | - Anastassia Rudik
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia.
| | - Alexander Dmitriev
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia.
| | - Alexey Lagunin
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia.
- Pirogov Russian National Research Medical University, 1 Ostrovityanova Str., 117997 Moscow, Russia.
| | - Dmitry Filimonov
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia.
| | - Vladimir Poroikov
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia.
| |
Collapse
|
24
|
Rudik AV, Dmitriev AV, Bezhentsev VM, Lagunin AA, Filimonov DA, Poroikov VV. Prediction of metabolites of epoxidation reaction in MetaTox. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2017; 28:833-842. [PMID: 29157013 DOI: 10.1080/1062936x.2017.1399165] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 10/27/2017] [Indexed: 06/07/2023]
Abstract
Biotransformation is a process of the chemical modifications which may lead to the reactive metabolites, in particular the epoxides. Epoxide reactive metabolites may cause the toxic effects. The prediction of such metabolites is important for drug development and ecotoxicology studies. Epoxides are formed by some oxidation reactions, usually catalysed by cytochromes P450, and represent a large class of three-membered cyclic ethers. Identification of molecules, which may be epoxidized, and indication of the specific location of epoxide functional group (which is called SOE - site of epoxidation) are important for prediction of epoxide metabolites. Datasets from 355 molecules and 615 reactions were created for training and validation. The prediction of SOE is based on a combination of LMNA (Labelled Multilevel Neighbourhood of Atom) descriptors and Bayesian-like algorithm implemented in PASS software and MetaTox web-service. The average invariant accuracy of prediction (AUC) calculated in leave-one-out and 20-fold cross-validation procedures is 0.9. Prediction of epoxide formation based on the created SAR model is included as the component of MetaTox web-service ( http://www.way2drug.com/mg ).
Collapse
Affiliation(s)
- A V Rudik
- a Institute of Biomedical Chemistry (IBMC) , Moscow , Russia
| | - A V Dmitriev
- a Institute of Biomedical Chemistry (IBMC) , Moscow , Russia
| | - V M Bezhentsev
- a Institute of Biomedical Chemistry (IBMC) , Moscow , Russia
| | - A A Lagunin
- a Institute of Biomedical Chemistry (IBMC) , Moscow , Russia
- b Medico-biological Faculty , Pirogov Russian National Research Medical University , Moscow , Russia
| | - D A Filimonov
- a Institute of Biomedical Chemistry (IBMC) , Moscow , Russia
| | - V V Poroikov
- a Institute of Biomedical Chemistry (IBMC) , Moscow , Russia
| |
Collapse
|
25
|
Dmitriev A, Rudik A, Filimonov D, Lagunin A, Pogodin P, Dubovskaja V, Bezhentsev V, Ivanov S, Druzhilovsky D, Tarasova O, Poroikov V. Integral estimation of xenobiotics’ toxicity with regard to their metabolism in human organism. PURE APPL CHEM 2017. [DOI: 10.1515/pac-2016-1205] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
AbstractToxicity and severe adverse effects are the primary cause of drug-candidate failures at the late stages of preclinical and clinical trials. Since most xenobiotics undergo biotransformations, their interaction with human organism reveals the effects produced by parent compounds and all metabolites. To increase the chances of successful drug development, estimation of the entire toxicity for drug substance and its metabolites is necessary for filtering out the potentially toxic compounds. We proposed the computational approach to the integral evaluation of xenobiotics’ toxicity based on the structural formula of the drug-like compound. In the framework of this study, the consensus QSAR model was developed based on the analysis of over 3000 compounds with information about their rat acute toxicity for intravenous route of administration. Four different numerical methods, estimating the integral toxicity, were proposed, and their comparative performance was studied using the external evaluation set consisting of 37 structures of drugs and 200 their metabolites. It was shown that, on the average, the best correspondence between the predicted and published data is obtained using the method that takes into account the estimated characteristics for both the parent compound and its most toxic metabolite.
Collapse
Affiliation(s)
- Alexander Dmitriev
- Institute of Biomedical Chemistry, 10 bldg. 7 Pogodinskaya Str., Moscow 119121, Russia
| | - Anastasia Rudik
- Institute of Biomedical Chemistry, 10 bldg. 7 Pogodinskaya Str., Moscow 119121, Russia
| | - Dmitry Filimonov
- Institute of Biomedical Chemistry, 10 bldg. 7 Pogodinskaya Str., Moscow 119121, Russia
| | - Alexey Lagunin
- Institute of Biomedical Chemistry, 10 bldg. 7 Pogodinskaya Str., Moscow 119121, Russia
- Medico-biological Faculty, Pirogov Russian National Research Medical University, 1 Ostrovityanova Str., Moscow 117997, Russia
| | - Pavel Pogodin
- Institute of Biomedical Chemistry, 10 bldg. 7 Pogodinskaya Str., Moscow 119121, Russia
| | - Varvara Dubovskaja
- Institute of Biomedical Chemistry, 10 bldg. 7 Pogodinskaya Str., Moscow 119121, Russia
| | - Vladislav Bezhentsev
- Institute of Biomedical Chemistry, 10 bldg. 7 Pogodinskaya Str., Moscow 119121, Russia
| | - Sergey Ivanov
- Institute of Biomedical Chemistry, 10 bldg. 7 Pogodinskaya Str., Moscow 119121, Russia
- Medico-biological Faculty, Pirogov Russian National Research Medical University, 1 Ostrovityanova Str., Moscow 117997, Russia
| | - Dmitry Druzhilovsky
- Institute of Biomedical Chemistry, 10 bldg. 7 Pogodinskaya Str., Moscow 119121, Russia
| | - Olga Tarasova
- Institute of Biomedical Chemistry, 10 bldg. 7 Pogodinskaya Str., Moscow 119121, Russia
| | - Vladimir Poroikov
- Institute of Biomedical Chemistry, 10 bldg. 7 Pogodinskaya Str., Moscow 119121, Russia
| |
Collapse
|
26
|
Dixit VA, Lal LA, Agrawal SR. Recent advances in the prediction of non‐
CYP450
‐mediated drug metabolism. WILEY INTERDISCIPLINARY REVIEWS-COMPUTATIONAL MOLECULAR SCIENCE 2017. [DOI: 10.1002/wcms.1323] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Vaibhav A. Dixit
- Department of Pharmaceutical Chemistry, School of Pharmacy & Technology Management (SPTM)Shri Vile Parle Kelavani Mandal's (SVKM's), Narsee Monjee Institute of Management Studies (NMIMS)ShirpurIndia
| | - L. Arun Lal
- Department of Pharmaceutical Chemistry, School of Pharmacy & Technology Management (SPTM)Shri Vile Parle Kelavani Mandal's (SVKM's), Narsee Monjee Institute of Management Studies (NMIMS)ShirpurIndia
| | - Simran R. Agrawal
- Department of Pharmaceutical Chemistry, School of Pharmacy & Technology Management (SPTM)Shri Vile Parle Kelavani Mandal's (SVKM's), Narsee Monjee Institute of Management Studies (NMIMS)ShirpurIndia
| |
Collapse
|
27
|
Rudik AV, Bezhentsev VM, Dmitriev AV, Druzhilovskiy DS, Lagunin AA, Filimonov DA, Poroikov VV. MetaTox: Web Application for Predicting Structure and Toxicity of Xenobiotics’ Metabolites. J Chem Inf Model 2017; 57:638-642. [DOI: 10.1021/acs.jcim.6b00662] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Anastasia V. Rudik
- Institute of Biomedical Chemistry (IBMC), 10 Bldg. 8, Pogodinskaya Str., Moscow, 119121, Russia
| | - Vladislav M. Bezhentsev
- Institute of Biomedical Chemistry (IBMC), 10 Bldg. 8, Pogodinskaya Str., Moscow, 119121, Russia
| | - Alexander V. Dmitriev
- Institute of Biomedical Chemistry (IBMC), 10 Bldg. 8, Pogodinskaya Str., Moscow, 119121, Russia
| | - Dmitry S. Druzhilovskiy
- Institute of Biomedical Chemistry (IBMC), 10 Bldg. 8, Pogodinskaya Str., Moscow, 119121, Russia
| | - Alexey A. Lagunin
- Institute of Biomedical Chemistry (IBMC), 10 Bldg. 8, Pogodinskaya Str., Moscow, 119121, Russia
- Medico-biological
Faculty, Pirogov Russian National Research Medical University, 1
Ostrovityanova Str., Moscow, 117997, Russia
| | - Dmitry A. Filimonov
- Institute of Biomedical Chemistry (IBMC), 10 Bldg. 8, Pogodinskaya Str., Moscow, 119121, Russia
| | - Vladimir V. Poroikov
- Institute of Biomedical Chemistry (IBMC), 10 Bldg. 8, Pogodinskaya Str., Moscow, 119121, Russia
| |
Collapse
|
28
|
Fu CW, Lin TH. Predicting the Metabolic Sites by Flavin-Containing Monooxygenase on Drug Molecules Using SVM Classification on Computed Quantum Mechanics and Circular Fingerprints Molecular Descriptors. PLoS One 2017; 12:e0169910. [PMID: 28072829 PMCID: PMC5224990 DOI: 10.1371/journal.pone.0169910] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 12/22/2016] [Indexed: 01/02/2023] Open
Abstract
As an important enzyme in Phase I drug metabolism, the flavin-containing monooxygenase (FMO) also metabolizes some xenobiotics with soft nucleophiles. The site of metabolism (SOM) on a molecule is the site where the metabolic reaction is exerted by an enzyme. Accurate prediction of SOMs on drug molecules will assist the search for drug leads during the optimization process. Here, some quantum mechanics features such as the condensed Fukui function and attributes from circular fingerprints (called Molprint2D) are computed and classified using the support vector machine (SVM) for predicting some potential SOMs on a series of drugs that can be metabolized by FMO enzymes. The condensed Fukui function fA- representing the nucleophilicity of central atom A and the attributes from circular fingerprints accounting the influence of neighbors on the central atom. The total number of FMO substrates and non-substrates collected in the study is 85 and they are equally divided into the training and test sets with each carrying roughly the same number of potential SOMs. However, only N-oxidation and S-oxidation features were considered in the prediction since the available C-oxidation data was scarce. In the training process, the LibSVM package of WEKA package and the option of 10-fold cross validation are employed. The prediction performance on the test set evaluated by accuracy, Matthews correlation coefficient and area under ROC curve computed are 0.829, 0.659, and 0.877 respectively. This work reveals that the SVM model built can accurately predict the potential SOMs for drug molecules that are metabolizable by the FMO enzymes.
Collapse
Affiliation(s)
- Chien-wei Fu
- Department of Pharmacy, National Taiwan University Hospital Hsin-Chu Branch, Institute of Molecular Medicine and Department of Life Science, National Tsing Hua University, HsinChu, Taiwan, ROC
| | - Thy-Hou Lin
- Department of Pharmacy, National Taiwan University Hospital Hsin-Chu Branch, Institute of Molecular Medicine and Department of Life Science, National Tsing Hua University, HsinChu, Taiwan, ROC
- * E-mail:
| |
Collapse
|
29
|
Danielson ML, Hu B, Shen J, Desai PV. In Silico ADME Techniques Used in Early-Phase Drug Discovery. TRANSLATING MOLECULES INTO MEDICINES 2017. [DOI: 10.1007/978-3-319-50042-3_4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
30
|
Rudik AV, Dmitriev AV, Lagunin AA, Filimonov DA, Poroikov VV. Prediction of reacting atoms for the major biotransformation reactions of organic xenobiotics. J Cheminform 2016; 8:68. [PMID: 27994650 PMCID: PMC5127045 DOI: 10.1186/s13321-016-0183-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 11/20/2016] [Indexed: 11/10/2022] Open
Abstract
Background The knowledge of drug metabolite structures is essential at the early stage of drug discovery to understand the potential liabilities and risks connected with biotransformation. The determination of the site of a molecule at which a particular metabolic reaction occurs could be used as a starting point for metabolite identification. The prediction of the site of metabolism does not always correspond to the particular atom that is modified by the enzyme but rather is often associated with a group of atoms. To overcome this problem, we propose to operate with the term “reacting atom”, corresponding to a single atom in the substrate that is modified during the biotransformation reaction. The prediction of the reacting atom(s) in a molecule for the major classes of biotransformation reactions is necessary to generate drug metabolites. Results Substrates of the major human cytochromes P450 and UDP-glucuronosyltransferases from the Biovia Metabolite database were divided into nine groups according to their reaction classes, which are aliphatic and aromatic hydroxylation, N- and O-glucuronidation, N-, S- and C-oxidation, and N- and O-dealkylation. Each training set consists of positive and negative examples of structures with one labelled atom. In the positive examples, the labelled atom is the reacting atom of a particular reaction that changed adjacency. Negative examples represent non-reacting atoms of a particular reaction. We used Labelled Multilevel Neighbourhoods of Atoms descriptors for the designation of reacting atoms. A Bayesian-like algorithm was applied to estimate the structure–activity relationships. The average invariant accuracy of prediction obtained in leave-one-out and 20-fold cross-validation procedures for five human isoforms of cytochrome P450 and all isoforms of UDP-glucuronosyltransferase varies from 0.86 to 0.99 (0.96 on average). Conclusions We report that reacting atoms may be predicted with reasonable accuracy for the major classes of metabolic reactions—aliphatic and aromatic hydroxylation, N- and O-glucuronidation, N-, S- and C-oxidation, and N- and O-dealkylation. The proposed method is implemented as a freely available web service at http://www.way2drug.com/RA and may be used for the prediction of the most probable biotransformation reaction(s) and the appropriate reacting atoms in drug-like compounds.. ![]() Electronic supplementary material The online version of this article (doi:10.1186/s13321-016-0183-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anastasia V Rudik
- Laboratory for Structure-Function Based Drug Design, Institute of Biomedical Chemistry, 10/8 Pogodinskaya Str., Moscow, Russia 119121
| | - Alexander V Dmitriev
- Laboratory for Structure-Function Based Drug Design, Institute of Biomedical Chemistry, 10/8 Pogodinskaya Str., Moscow, Russia 119121
| | - Alexey A Lagunin
- Laboratory for Structure-Function Based Drug Design, Institute of Biomedical Chemistry, 10/8 Pogodinskaya Str., Moscow, Russia 119121 ; Medico-Biological Faculty, Pirogov Russian National Research Medical University, 1 Ostrovityanova Str., Moscow, Russia 117997
| | - Dmitry A Filimonov
- Laboratory for Structure-Function Based Drug Design, Institute of Biomedical Chemistry, 10/8 Pogodinskaya Str., Moscow, Russia 119121
| | - Vladimir V Poroikov
- Laboratory for Structure-Function Based Drug Design, Institute of Biomedical Chemistry, 10/8 Pogodinskaya Str., Moscow, Russia 119121
| |
Collapse
|
31
|
Druzhilovskiy DS, Rudik AV, Filimonov DA, Lagunin AA, Gloriozova TA, Poroikov VV. Online resources for the prediction of biological activity of organic compounds. Russ Chem Bull 2016. [DOI: 10.1007/s11172-016-1310-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
32
|
Spjuth O, Rydberg P, Willighagen EL, Evelo CT, Jeliazkova N. XMetDB: an open access database for xenobiotic metabolism. J Cheminform 2016; 8:47. [PMID: 27651835 PMCID: PMC5025591 DOI: 10.1186/s13321-016-0161-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 09/04/2016] [Indexed: 12/17/2022] Open
Abstract
Xenobiotic metabolism is an active research topic but the limited amount of openly available high-quality biotransformation data constrains predictive modeling. Current database often default to commonly available information: which enzyme metabolizes a compound, but neither experimental conditions nor the atoms that undergo metabolization are captured. We present XMetDB, an open access database for drugs and other xenobiotics and their respective metabolites. The database contains chemical structures of xenobiotic biotransformations with substrate atoms annotated as reaction centra, the resulting product formed, and the catalyzing enzyme, type of experiment, and literature references. Associated with the database is a web interface for the submission and retrieval of experimental metabolite data for drugs and other xenobiotics in various formats, and a web API for programmatic access is also available. The database is open for data deposition, and a curation scheme is in place for quality control. An extensive guide on how to enter experimental data into is available from the XMetDB wiki. XMetDB formalizes how biotransformation data should be reported, and the openly available systematically labeled data is a big step forward towards better models for predictive metabolism.
Collapse
Affiliation(s)
- Ola Spjuth
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, 75124 Uppsala, Sweden
| | - Patrik Rydberg
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Egon L Willighagen
- Department of Bioinformatics - BiGCaT, Maastricht University, P.O. Box 616, UNS50 Box 19, 6200 MD Maastricht, The Netherlands
| | - Chris T Evelo
- Department of Bioinformatics - BiGCaT, Maastricht University, P.O. Box 616, UNS50 Box 19, 6200 MD Maastricht, The Netherlands
| | | |
Collapse
|
33
|
Abstract
In recent decades, in silico absorption, distribution, metabolism, excretion (ADME), and toxicity (T) modelling as a tool for rational drug design has received considerable attention from pharmaceutical scientists, and various ADME/T-related prediction models have been reported. The high-throughput and low-cost nature of these models permits a more streamlined drug development process in which the identification of hits or their structural optimization can be guided based on a parallel investigation of bioavailability and safety, along with activity. However, the effectiveness of these tools is highly dependent on their capacity to cope with needs at different stages, e.g. their use in candidate selection has been limited due to their lack of the required predictability. For some events or endpoints involving more complex mechanisms, the current in silico approaches still need further improvement. In this review, we will briefly introduce the development of in silico models for some physicochemical parameters, ADME properties and toxicity evaluation, with an emphasis on the modelling approaches thereof, their application in drug discovery, and the potential merits or deficiencies of these models. Finally, the outlook for future ADME/T modelling based on big data analysis and systems sciences will be discussed.
Collapse
|
34
|
Olsen L, Oostenbrink C, Jørgensen FS. Prediction of cytochrome P450 mediated metabolism. Adv Drug Deliv Rev 2015; 86:61-71. [PMID: 25958010 DOI: 10.1016/j.addr.2015.04.020] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 03/30/2015] [Accepted: 04/27/2015] [Indexed: 10/23/2022]
Abstract
Cytochrome P450 enzymes (CYPs) form one of the most important enzyme families involved in the metabolism of xenobiotics. CYPs comprise many isoforms, which catalyze a wide variety of reactions, and potentially, a large number of different metabolites can be formed. However, it is often hard to rationalize what metabolites these enzymes generate. In recent years, many different in silico approaches have been developed to predict binding or regioselective product formation for the different CYP isoforms. These comprise ligand-based methods that are trained on experimental CYP data and structure-based methods that consider how the substrate is oriented in the active site or/and how reactive the part of the substrate that is accessible to the heme group is. We will review key aspects for various approaches that are available to predict binding and site of metabolism (SOM), what outcome can be expected from the predictions, and how they could potentially be improved.
Collapse
|
35
|
Raunio H, Kuusisto M, Juvonen RO, Pentikäinen OT. Modeling of interactions between xenobiotics and cytochrome P450 (CYP) enzymes. Front Pharmacol 2015; 6:123. [PMID: 26124721 PMCID: PMC4464169 DOI: 10.3389/fphar.2015.00123] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 05/29/2015] [Indexed: 01/01/2023] Open
Abstract
The adverse effects to humans and environment of only few chemicals are well known. Absorption, distribution, metabolism, and excretion (ADME) are the steps of pharmaco/toxicokinetics that determine the internal dose of chemicals to which the organism is exposed. Of all the xenobiotic-metabolizing enzymes, the cytochrome P450 (CYP) enzymes are the most important due to their abundance and versatility. Reactions catalyzed by CYPs usually turn xenobiotics to harmless and excretable metabolites, but sometimes an innocuous xenobiotic is transformed into a toxic metabolite. Data on ADME and toxicity properties of compounds are increasingly generated using in vitro and modeling (in silico) tools. Both physics-based and empirical modeling approaches are used. Numerous ligand-based and target-based as well as combined modeling methods have been employed to evaluate determinants of CYP ligand binding as well as predicting sites of metabolism and inhibition characteristics of test molecules. In silico prediction of CYP–ligand interactions have made crucial contributions in understanding (1) determinants of CYP ligand binding recognition and affinity; (2) prediction of likely metabolites from substrates; (3) prediction of inhibitors and their inhibition potency. Truly predictive models of toxic outcomes cannot be created without incorporating metabolic characteristics; in silico methods help producing such information and filling gaps in experimentally derived data. Currently modeling methods are not mature enough to replace standard in vitro and in vivo approaches, but they are already used as an important component in risk assessment of drugs and other chemicals.
Collapse
Affiliation(s)
- Hannu Raunio
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland Kuopio, Finland
| | - Mira Kuusisto
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland Kuopio, Finland ; Computational Bioscience Laboratory, Department of Biological and Environmental Science, Nanoscience Center, University of Jyväskylä Jyväskylä, Finland
| | - Risto O Juvonen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland Kuopio, Finland
| | - Olli T Pentikäinen
- Computational Bioscience Laboratory, Department of Biological and Environmental Science, Nanoscience Center, University of Jyväskylä Jyväskylä, Finland
| |
Collapse
|
36
|
Zaretzki JM, Browning MR, Hughes TB, Swamidass SJ. Extending P450 site-of-metabolism models with region-resolution data. Bioinformatics 2015; 31:1966-73. [DOI: 10.1093/bioinformatics/btv100] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 02/11/2015] [Indexed: 12/18/2022] Open
|
37
|
Rudik A, Dmitriev A, Lagunin A, Filimonov D, Poroikov V. SOMP: web server for in silico prediction of sites of metabolism for drug-like compounds. Bioinformatics 2015; 31:2046-8. [PMID: 25777527 DOI: 10.1093/bioinformatics/btv087] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 02/07/2015] [Indexed: 11/12/2022] Open
Abstract
UNLABELLED A new freely available web server site of metabolism predictor to predict the sites of metabolism (SOM) based on the structural formula of chemicals has been developed. It is based on the analyses of 'structure-SOM' relationships using a Bayesian approach and labelled multilevel neighbourhoods of atoms descriptors to represent the structures of over 1000 metabolized xenobiotics. The server allows predicting SOMs that are catalysed by 1A2, 2C9, 2C19, 2D6 and 3A4 isoforms of cytochrome P450 and enzymes of the UDP-glucuronosyltransferase family. The average invariant accuracy of prediction that was calculated for the training sets (using leave-one-out cross-validation) and evaluation sets is 0.9 and 0.95, respectively. AVAILABILITY AND IMPLEMENTATION Freely available on the web at http://www.way2drug.com/SOMP.
Collapse
Affiliation(s)
- Anastasia Rudik
- Laboratory for Structure-Function Based Drug Design, Institute of Biomedical Chemistry, 10/8 Pogodinskaya Str., Moscow, 119121, Russia and Medicobiological Faculty, Pirogov Russian National Research Medical University, 1 Ostrovityanova Str., Moscow, 117997, Russia
| | - Alexander Dmitriev
- Laboratory for Structure-Function Based Drug Design, Institute of Biomedical Chemistry, 10/8 Pogodinskaya Str., Moscow, 119121, Russia and Medicobiological Faculty, Pirogov Russian National Research Medical University, 1 Ostrovityanova Str., Moscow, 117997, Russia
| | - Alexey Lagunin
- Laboratory for Structure-Function Based Drug Design, Institute of Biomedical Chemistry, 10/8 Pogodinskaya Str., Moscow, 119121, Russia and Medicobiological Faculty, Pirogov Russian National Research Medical University, 1 Ostrovityanova Str., Moscow, 117997, Russia Laboratory for Structure-Function Based Drug Design, Institute of Biomedical Chemistry, 10/8 Pogodinskaya Str., Moscow, 119121, Russia and Medicobiological Faculty, Pirogov Russian National Research Medical University, 1 Ostrovityanova Str., Moscow, 117997, Russia
| | - Dmitry Filimonov
- Laboratory for Structure-Function Based Drug Design, Institute of Biomedical Chemistry, 10/8 Pogodinskaya Str., Moscow, 119121, Russia and Medicobiological Faculty, Pirogov Russian National Research Medical University, 1 Ostrovityanova Str., Moscow, 117997, Russia
| | - Vladimir Poroikov
- Laboratory for Structure-Function Based Drug Design, Institute of Biomedical Chemistry, 10/8 Pogodinskaya Str., Moscow, 119121, Russia and Medicobiological Faculty, Pirogov Russian National Research Medical University, 1 Ostrovityanova Str., Moscow, 117997, Russia Laboratory for Structure-Function Based Drug Design, Institute of Biomedical Chemistry, 10/8 Pogodinskaya Str., Moscow, 119121, Russia and Medicobiological Faculty, Pirogov Russian National Research Medical University, 1 Ostrovityanova Str., Moscow, 117997, Russia
| |
Collapse
|
38
|
Sheng Y, Chen Y, Wang L, Liu G, Li W, Tang Y. Effects of protein flexibility on the site of metabolism prediction for CYP2A6 substrates. J Mol Graph Model 2014; 54:90-9. [DOI: 10.1016/j.jmgm.2014.09.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 09/25/2014] [Accepted: 09/30/2014] [Indexed: 12/01/2022]
|
39
|
Tyzack JD, Mussa HY, Williamson MJ, Kirchmair J, Glen RC. Cytochrome P450 site of metabolism prediction from 2D topological fingerprints using GPU accelerated probabilistic classifiers. J Cheminform 2014; 6:29. [PMID: 24959208 PMCID: PMC4047555 DOI: 10.1186/1758-2946-6-29] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 05/12/2014] [Indexed: 02/02/2023] Open
Abstract
Background The prediction of sites and products of metabolism in xenobiotic compounds is key to the development of new chemical entities, where screening potential metabolites for toxicity or unwanted side-effects is of crucial importance. In this work 2D topological fingerprints are used to encode atomic sites and three probabilistic machine learning methods are applied: Parzen-Rosenblatt Window (PRW), Naive Bayesian (NB) and a novel approach called RASCAL (Random Attribute Subsampling Classification ALgorithm). These are implemented by randomly subsampling descriptor space to alleviate the problem often suffered by data mining methods of having to exactly match fingerprints, and in the case of PRW by measuring a distance between feature vectors rather than exact matching. The classifiers have been implemented in CUDA/C++ to exploit the parallel architecture of graphical processing units (GPUs) and is freely available in a public repository. Results It is shown that for PRW a SoM (Site of Metabolism) is identified in the top two predictions for 85%, 91% and 88% of the CYP 3A4, 2D6 and 2C9 data sets respectively, with RASCAL giving similar performance of 83%, 91% and 88%, respectively. These results put PRW and RASCAL performance ahead of NB which gave a much lower classification performance of 51%, 73% and 74%, respectively. Conclusions 2D topological fingerprints calculated to a bond depth of 4-6 contain sufficient information to allow the identification of SoMs using classifiers based on relatively small data sets. Thus, the machine learning methods outlined in this paper are conceptually simpler and more efficient than other methods tested and the use of simple topological descriptors derived from 2D structure give results competitive with other approaches using more expensive quantum chemical descriptors. The descriptor space subsampling approach and ensemble methodology allow the methods to be applied to molecules more distant from the training data where data mining would be more likely to fail due to the lack of common fingerprints. The RASCAL algorithm is shown to give equivalent classification performance to PRW but at lower computational expense allowing it to be applied more efficiently in the ensemble scheme.
Collapse
Affiliation(s)
- Jonathan D Tyzack
- Unilever Centre for Molecular Science Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, UK
| | - Hamse Y Mussa
- Unilever Centre for Molecular Science Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, UK
| | - Mark J Williamson
- Unilever Centre for Molecular Science Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, UK
| | - Johannes Kirchmair
- ETH Zurich, Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, HCI G 474.2, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Robert C Glen
- Unilever Centre for Molecular Science Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, UK
| |
Collapse
|