1
|
Li Y, Xu QW, Jian GL, Zhang XL, Wang H. Improved and Interpretable Prediction of Cytochrome P450-Mediated Metabolism by Molecule-Level Graph Modeling and Subgraph Information Bottlenecks. J Chem Inf Model 2024; 64:9487-9500. [PMID: 39602328 DOI: 10.1021/acs.jcim.4c01632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Accurately identifying sites of metabolism (SoM) mediated by cytochrome P450 (CYP) enzymes, which are responsible for drug metabolism in the body, is critical in the early stage of drug discovery and development. Current computational methods for CYP-mediated SoM prediction face several challenges, including limitations to traditional machine learning models at the atomic level, heavy reliance on complex feature engineering, and the lack of interpretability relevant to medicinal chemistry. Here, we propose GraphCySoM, a novel molecule-level modeling approach based on graph neural networks, utilizing lightweight features and interpretable annotations on substructures, to effectively and interpretably predict CYP-mediated SoM. Unlike computationally expensive atomic descriptors derived from resource-intensive chemistry or even quantum chemistry calculations, we emphasize that graph-based molecular modeling initialized solely with lightweight features enables the adaptive learning of molecular topology through message-passing mechanisms combined with various aggregation kernels. Extensive ablation experiments demonstrate that GraphCySoM significantly outperforms baseline models and achieves superior performance compared with competing methods while exhibiting advantages in computational efficiency. Moreover, the attention mechanism and subgraph information bottlenecks are incorporated to analyze node importance and feature significance, resulting in mining substructures associated with the SoM. To the best of our knowledge, this is the first comprehensive study of CYP-mediated SoM using molecule-level modeling and interpretable technology. Our method achieves new state-of-the-art performance and provides potential insights into the molecular and pharmacological mechanisms underlying drug metabolism catalyzed by CYP enzymes. All source files and trained models are freely available at https://github.com/liyigerry/GraphCySoM.
Collapse
Affiliation(s)
- Yi Li
- College of Mathematics and Computer Science, Dali University, Dali 671003, China
| | - Qin-Wei Xu
- College of Mathematics and Computer Science, Dali University, Dali 671003, China
| | - Guo-Lei Jian
- College of Mathematics and Computer Science, Dali University, Dali 671003, China
| | - Xiao-Ling Zhang
- College of Mathematics and Computer Science, Dali University, Dali 671003, China
| | - Hua Wang
- College of Metallurgy and Energy Engineering, Kunming University of Science and Technology, Kunming 650031, China
| |
Collapse
|
2
|
Sarkar B, Alam S, Rajib TK, Islam SS, Araf Y, Ullah MA. Identification of the most potent acetylcholinesterase inhibitors from plants for possible treatment of Alzheimer’s disease: a computational approach. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2021. [DOI: 10.1186/s43042-020-00127-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Being one of the rapidly growing dementia type diseases in the world, Alzheimer’s disease (AD) has gained much attention from researchers in the recent decades. Many hypotheses have been developed that describe different reasons for the development of AD. Among them, the cholinergic hypothesis depicts that the degradation of an important neurotransmitter, acetylcholine by the enzyme acetylcholinesterase (AChE), is responsible for the development of AD. Although, many anti-AChE drugs are already available in the market, their performance sometimes yields unexpected results. For this reason, research works are going on to find out potential anti-AChE agents both from natural and synthetic sources. In this study, 50 potential anti-AChE phytochemicals were analyzed using numerous tools of bioinformatics and in silico biology to find out the best possible anti-AChE agents among the selected 50 ligands through molecular docking, determination of the druglikeness properties, conducting the ADMET test, PASS and P450 site of metabolism prediction, and DFT calculations.
Result
The predictions of this study suggested that among the selected 50 ligands, bellidifolin, naringenin, apigenin, and coptisine were the 4 best compounds with quite similar and sound performance in most of the experiments.
Conclusion
In this study, bellidifolin, naringenin, apigenin, and coptisine were found to be the most effective agents for treating the AD targeting AChE. However, more in vivo and in vitro analyses are required to finally confirm the outcomes of this research.
Collapse
|
3
|
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
|
4
|
Sarkar B, Ullah MA, Islam SS, Rahman MH, Araf Y. Analysis of plant-derived phytochemicals as anti-cancer agents targeting cyclin dependent kinase-2, human topoisomerase IIa and vascular endothelial growth factor receptor-2. J Recept Signal Transduct Res 2020; 41:217-233. [PMID: 32787531 DOI: 10.1080/10799893.2020.1805628] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancer is caused by a variety of pathways, involving numerous types of enzymes. Among them three enzymes i.e. Cyclin-dependent kinase-2 (CDK-2), Human topoisomerase IIα, and Vascular Endothelial Growth Factor Receptor-2 (VEGFR-2) are three of the most common enzymes that are involved in the cancer development. Although many chemical drugs are already available in the market for cancer treatment, plant sources are known to contain a wide variety of agents that are proved to possess potential anticancer activity. In this experiment, total thirty phytochemicals were analyzed against the mentioned three enzymes using different tools of bioinformatics and in silico biology like molecular docking study, drug likeness property experiment, ADME/T test, PASS prediction, and P450 site of metabolism prediction as well as DFT calculation to determine the three best ligands among them that have the capability to inhibit the mentioned enzymes. From the experiment, Epigallocatechin gallate was found to be the best ligand to inhibit CDK-2, Daidzein showed the best inhibitory activities towards the Human topoisomerase IIα, and Quercetin was predicted to be the best agent against VEGFR-2. They were also predicted to be quite safe and effective agents to treat cancer. However, more in vivo and in vitro analyses are required to finally confirm their safety and efficacy in this regard.
Collapse
Affiliation(s)
- Bishajit Sarkar
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Jahangirnagar University, Savar, Dhaka, Bangladesh
| | - Md Asad Ullah
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Jahangirnagar University, Savar, Dhaka, Bangladesh
| | - Syed Sajidul Islam
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Jahangirnagar University, Savar, Dhaka, Bangladesh
| | - Md Hasanur Rahman
- Department of Biotechnology and Genetic Engineering, Faculty of Life Sciences, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Yusha Araf
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| |
Collapse
|
5
|
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
|
6
|
Xiong Y, Qiao Y, Kihara D, Zhang HY, Zhu X, Wei DQ. Survey of Machine Learning Techniques for Prediction of the Isoform Specificity of Cytochrome P450 Substrates. Curr Drug Metab 2019; 20:229-235. [PMID: 30338736 DOI: 10.2174/1389200219666181019094526] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 08/05/2018] [Accepted: 08/06/2018] [Indexed: 12/23/2022]
Abstract
Background:Determination or prediction of the Absorption, Distribution, Metabolism, and Excretion (ADME) properties of drug candidates and drug-induced toxicity plays crucial roles in drug discovery and development. Metabolism is one of the most complicated pharmacokinetic properties to be understood and predicted. However, experimental determination of the substrate binding, selectivity, sites and rates of metabolism is time- and recourse- consuming. In the phase I metabolism of foreign compounds (i.e., most of drugs), cytochrome P450 enzymes play a key role. To help develop drugs with proper ADME properties, computational models are highly desired to predict the ADME properties of drug candidates, particularly for drugs binding to cytochrome P450.Objective:This narrative review aims to briefly summarize machine learning techniques used in the prediction of the cytochrome P450 isoform specificity of drug candidates.Results:Both single-label and multi-label classification methods have demonstrated good performance on modelling and prediction of the isoform specificity of substrates based on their quantitative descriptors.Conclusion:This review provides a guide for researchers to develop machine learning-based methods to predict the cytochrome P450 isoform specificity of drug candidates.
Collapse
Affiliation(s)
- Yi Xiong
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yanhua Qiao
- School of Life Sciences, Anhui University, Hefei, Anhui 230601, China
| | - Daisuke Kihara
- Department of Biological Science, Purdue University, West Lafayette, IN 47907, United States
| | - Hui-Yuan Zhang
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiaolei Zhu
- School of Life Sciences, Anhui University, Hefei, Anhui 230601, China
| | - Dong-Qing Wei
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
7
|
Mazzolari A, Afzal AM, Pedretti A, Testa B, Vistoli G, Bender A. Prediction of UGT-mediated Metabolism Using the Manually Curated MetaQSAR Database. ACS Med Chem Lett 2019; 10:633-638. [PMID: 30996809 DOI: 10.1021/acsmedchemlett.8b00603] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/12/2019] [Indexed: 11/30/2022] Open
Abstract
Even though glucuronidations are the most frequent metabolic reactions of conjugation, both in quantitative and qualitative terms, they have rather seldom been investigated using computational approaches. To fill this gap, we have used the manually collected MetaQSAR metabolic reaction database to generate two models for the prediction of UGT-mediated metabolism, both based on molecular descriptors and implementing the Random Forest algorithm. The first model predicts the occurrence of the reaction and was internally validated with a Matthew correlation coefficient (MCC) of 0.76 and an area under the ROC curve (AUC) of 0.94, and further externally validated using a test set composed of 120 additional xenobiotics (MCC of 0.70 and AUC of 0.90). The second model distinguishes between O- and N-glucuronidations and was optimized by the random undersampling procedure to improve the predictive accuracy during the internal validation, with the recall measure of the minority class increasing from 0.55 to 0.78.
Collapse
Affiliation(s)
- Angelica Mazzolari
- Dipartimento di Scienze Farmaceutiche, Facoltà di Scienze del Farmaco, Università degli Studi di Milano, Via Mangiagalli, I-20133 Milano, Italy
| | - Avid M. Afzal
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, U.K
| | - Alessandro Pedretti
- Dipartimento di Scienze Farmaceutiche, Facoltà di Scienze del Farmaco, Università degli Studi di Milano, Via Mangiagalli, I-20133 Milano, Italy
| | | | - Giulio Vistoli
- Dipartimento di Scienze Farmaceutiche, Facoltà di Scienze del Farmaco, Università degli Studi di Milano, Via Mangiagalli, I-20133 Milano, Italy
| | - Andreas Bender
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, U.K
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Schurz A, Su BH, Tu YS, Lu TTY, Lin OA, Tseng YJ. G.A.M.E.: GPU-accelerated mixture elucidator. J Cheminform 2017; 9:50. [PMID: 29086161 PMCID: PMC5602814 DOI: 10.1186/s13321-017-0238-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 09/05/2017] [Indexed: 11/23/2022] Open
Abstract
GPU acceleration is useful in solving complex chemical information problems. Identifying unknown structures from the mass spectra of natural product mixtures has been a desirable yet unresolved issue in metabolomics. However, this elucidation process has been hampered by complex experimental data and the inability of instruments to completely separate different compounds. Fortunately, with current high-resolution mass spectrometry, one feasible strategy is to define this problem as extending a scaffold database with sidechains of different probabilities to match the high-resolution mass obtained from a high-resolution mass spectrum. By introducing a dynamic programming (DP) algorithm, it is possible to solve this NP-complete problem in pseudo-polynomial time. However, the running time of the DP algorithm grows by orders of magnitude as the number of mass decimal digits increases, thus limiting the boost in structural prediction capabilities. By harnessing the heavily parallel architecture of modern GPUs, we designed a “compute unified device architecture” (CUDA)-based GPU-accelerated mixture elucidator (G.A.M.E.) that considerably improves the performance of the DP, allowing up to five decimal digits for input mass data. As exemplified by four testing datasets with verified constitutions from natural products, G.A.M.E. allows for efficient and automatic structural elucidation of unknown mixtures for practical procedures.. ![]()
Collapse
Affiliation(s)
- Alioune Schurz
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1 Sec. 4, Roosevelt Road, Taipei, 106, Taiwan
| | - Bo-Han Su
- Department of Computer Science and Information Engineering, National Taiwan University, No. 1 Sec. 4, Roosevelt Road, Taipei, 106, Taiwan
| | - Yi-Shu Tu
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1 Sec. 4, Roosevelt Road, Taipei, 106, Taiwan
| | - Tony Tsung-Yu Lu
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1 Sec. 4, Roosevelt Road, Taipei, 106, Taiwan
| | - Olivia A Lin
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1 Sec. 4, Roosevelt Road, Taipei, 106, Taiwan
| | - Yufeng J Tseng
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1 Sec. 4, Roosevelt Road, Taipei, 106, Taiwan. .,Department of Computer Science and Information Engineering, National Taiwan University, No. 1 Sec. 4, Roosevelt Road, Taipei, 106, Taiwan. .,Drug Research Center, National Taiwan University College of Medicine, No. 1 Sec. 1, Jen Ai Rord, Taipei, 106, Taiwan.
| |
Collapse
|
11
|
Šícho M, de Bruyn Kops C, Stork C, Svozil D, Kirchmair J. FAME 2: Simple and Effective Machine Learning Model of Cytochrome P450 Regioselectivity. J Chem Inf Model 2017; 57:1832-1846. [DOI: 10.1021/acs.jcim.7b00250] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Martin Šícho
- Faculty
of Mathematics, Informatics and Natural Sciences, Department of Computer
Science, Center for Bioinformatics, Universität Hamburg, Hamburg, 20146, Germany
- CZ-OPENSCREEN:
National Infrastructure for Chemical Biology, Laboratory of Informatics
and Chemistry, Faculty of Chemical Technology, University of Chemistry and Technology Prague, 166 28 Prague 6, Czech Republic
| | - Christina de Bruyn Kops
- Faculty
of Mathematics, Informatics and Natural Sciences, Department of Computer
Science, Center for Bioinformatics, Universität Hamburg, Hamburg, 20146, Germany
| | - Conrad Stork
- Faculty
of Mathematics, Informatics and Natural Sciences, Department of Computer
Science, Center for Bioinformatics, Universität Hamburg, Hamburg, 20146, Germany
| | - Daniel Svozil
- CZ-OPENSCREEN:
National Infrastructure for Chemical Biology, Laboratory of Informatics
and Chemistry, Faculty of Chemical Technology, University of Chemistry and Technology Prague, 166 28 Prague 6, Czech Republic
| | - Johannes Kirchmair
- Faculty
of Mathematics, Informatics and Natural Sciences, Department of Computer
Science, Center for Bioinformatics, Universität Hamburg, Hamburg, 20146, Germany
| |
Collapse
|
12
|
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
|
13
|
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]
|
14
|
Dixit VA, Deshpande S. Advances in Computational Prediction of Regioselective and Isoform-Specific Drug Metabolism Catalyzed by CYP450s. ChemistrySelect 2016. [DOI: 10.1002/slct.201601051] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Vaibhav A. Dixit
- Department of Pharmaceutical Chemistry; School of Pharmacy and Technology Management (SPTM), Shri Vile Parle Kelavani Mandal's (SVKM's) Narsee Monjee Institute of Management Studies (NMIMS), Mukesh Patel Technology Park, Babulde, Bank of Tapi River; Mumbai-Agra Road Shirpur, Dist. Dhule−425405 India
| | - Shirish Deshpande
- Department of Pharmaceutical Chemistry; School of Pharmacy and Technology Management (SPTM), Shri Vile Parle Kelavani Mandal's (SVKM's) Narsee Monjee Institute of Management Studies (NMIMS), Mukesh Patel Technology Park, Babulde, Bank of Tapi River; Mumbai-Agra Road Shirpur, Dist. Dhule−425405 India
| |
Collapse
|
15
|
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
|
16
|
Developing a Physiologically-Based Pharmacokinetic Model Knowledgebase in Support of Provisional Model Construction. PLoS Comput Biol 2016; 12:e1004495. [PMID: 26871706 PMCID: PMC4752336 DOI: 10.1371/journal.pcbi.1004495] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 08/03/2015] [Indexed: 11/19/2022] Open
Abstract
Developing physiologically-based pharmacokinetic (PBPK) models for chemicals can be resource-intensive, as neither chemical-specific parameters nor in vivo pharmacokinetic data are easily available for model construction. Previously developed, well-parameterized, and thoroughly-vetted models can be a great resource for the construction of models pertaining to new chemicals. A PBPK knowledgebase was compiled and developed from existing PBPK-related articles and used to develop new models. From 2,039 PBPK-related articles published between 1977 and 2013, 307 unique chemicals were identified for use as the basis of our knowledgebase. Keywords related to species, gender, developmental stages, and organs were analyzed from the articles within the PBPK knowledgebase. A correlation matrix of the 307 chemicals in the PBPK knowledgebase was calculated based on pharmacokinetic-relevant molecular descriptors. Chemicals in the PBPK knowledgebase were ranked based on their correlation toward ethylbenzene and gefitinib. Next, multiple chemicals were selected to represent exact matches, close analogues, or non-analogues of the target case study chemicals. Parameters, equations, or experimental data relevant to existing models for these chemicals and their analogues were used to construct new models, and model predictions were compared to observed values. This compiled knowledgebase provides a chemical structure-based approach for identifying PBPK models relevant to other chemical entities. Using suitable correlation metrics, we demonstrated that models of chemical analogues in the PBPK knowledgebase can guide the construction of PBPK models for other chemicals.
Collapse
|
17
|
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
|
18
|
Fuchs JE, Bender A, Glen RC. Cheminformatics Research at the Unilever Centre for Molecular Science Informatics Cambridge. Mol Inform 2015; 34:626-633. [PMID: 26435758 PMCID: PMC4583778 DOI: 10.1002/minf.201400166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 12/16/2014] [Indexed: 11/12/2022]
Abstract
The Centre for Molecular Informatics, formerly Unilever Centre for Molecular Science Informatics (UCMSI), at the University of Cambridge is a world-leading driving force in the field of cheminformatics. Since its opening in 2000 more than 300 scientific articles have fundamentally changed the field of molecular informatics. The Centre has been a key player in promoting open chemical data and semantic access. Though mainly focussing on basic research, close collaborations with industrial partners ensured real world feedback and access to high quality molecular data. A variety of tools and standard protocols have been developed and are ubiquitous in the daily practice of cheminformatics. Here, we present a retrospective of cheminformatics research performed at the UCMSI, thereby highlighting historical and recent trends in the field as well as indicating future directions.
Collapse
Affiliation(s)
- Julian E Fuchs
- Centre for Molecular Informatics, Department of Chemistry, University of CambridgeLensfield Road, Cambridge CB2 1EW, UK phone/fax: +44 (0)1223 336472/+44 (0)1223 763076
| | - Andreas Bender
- Centre for Molecular Informatics, Department of Chemistry, University of CambridgeLensfield Road, Cambridge CB2 1EW, UK phone/fax: +44 (0)1223 336472/+44 (0)1223 763076
| | - Robert C Glen
- Centre for Molecular Informatics, Department of Chemistry, University of CambridgeLensfield Road, Cambridge CB2 1EW, UK phone/fax: +44 (0)1223 336472/+44 (0)1223 763076
| |
Collapse
|
19
|
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
|
20
|
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
|
21
|
Mussa HY, Marcus D, Mitchell JBO, Glen RC. Verifying the fully "Laplacianised" posterior Naïve Bayesian approach and more. J Cheminform 2015; 7:27. [PMID: 26075027 PMCID: PMC4464057 DOI: 10.1186/s13321-015-0075-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 05/12/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In a recent paper, Mussa, Mitchell and Glen (MMG) have mathematically demonstrated that the "Laplacian Corrected Modified Naïve Bayes" (LCMNB) algorithm can be viewed as a variant of the so-called Standard Naïve Bayes (SNB) scheme, whereby the role played by absence of compound features in classifying/assigning the compound to its appropriate class is ignored. MMG have also proffered guidelines regarding the conditions under which this omission may hold. Utilising three data sets, the present paper examines the validity of these guidelines in practice. The paper also extends MMG's work and introduces a new version of the SNB classifier: "Tapered Naïve Bayes" (TNB). TNB does not discard the role of absence of a feature out of hand, nor does it fully consider its role. Hence, TNB encapsulates both SNB and LCMNB. RESULTS LCMNB, SNB and TNB performed differently on classifying 4,658, 5,031 and 1,149 ligands (all chosen from the ChEMBL Database) distributed over 31 enzymes, 23 membrane receptors, and one ion-channel, four transporters and one transcription factor as their target proteins. When the number of features utilised was equal to or smaller than the "optimal" number of features for a given data set, SNB classifiers systematically gave better classification results than those yielded by LCMNB classifiers. The opposite was true when the number of features employed was markedly larger than the "optimal" number of features for this data set. Nonetheless, these LCMNB performances were worse than the classification performance achieved by SNB when the "optimal" number of features for the data set was utilised. TNB classifiers systematically outperformed both SNB and LCMNB classifiers. CONCLUSIONS The classification results obtained in this study concur with the mathematical based guidelines given in MMG's paper-that is, ignoring the role of absence of a feature out of hand does not necessarily improve classification performance of the SNB approach; if anything, it could make the performance of the SNB method worse. The results obtained also lend support to the rationale, on which the TNB algorithm rests: handled judiciously, taking into account absence of features can enhance (not impair) the discriminatory classification power of the SNB approach.
Collapse
Affiliation(s)
- Hamse Y Mussa
- />Department of Chemistry, Centre for Molecular Informatics, Lensfield Road, Cambridge, England CB2 1EW UK
- />EaStCHEM School of Chemistry and Biomedical Sciences Research Complex, University of St Andrews, North Haugh, St Andrews, Scotland KY16 9ST UK
| | - David Marcus
- />European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Wellcome Trust Genome Campus, Hinxton, Cambridge, England CB10 1SD UK
| | - John B O Mitchell
- />EaStCHEM School of Chemistry and Biomedical Sciences Research Complex, University of St Andrews, North Haugh, St Andrews, Scotland KY16 9ST UK
| | - Robert C Glen
- />Department of Chemistry, Centre for Molecular Informatics, Lensfield Road, Cambridge, England CB2 1EW UK
| |
Collapse
|
22
|
Zaretzki J, Boehm KM, Swamidass SJ. Improved Prediction of CYP-Mediated Metabolism with Chemical Fingerprints. J Chem Inf Model 2015; 55:972-82. [DOI: 10.1021/ci5005652] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Jed Zaretzki
- Department of Pathology and
Immunology, Washington University School of Medicine, Campus Box
1097 Whitaker Hall, St. Louis, Missouri 63130, United States
| | - Kevin M. Boehm
- Department of Pathology and
Immunology, Washington University School of Medicine, Campus Box
1097 Whitaker Hall, St. Louis, Missouri 63130, United States
| | - S. Joshua Swamidass
- Department of Pathology and
Immunology, Washington University School of Medicine, Campus Box
1097 Whitaker Hall, St. Louis, Missouri 63130, United States
| |
Collapse
|
23
|
von Grafenstein S, Fuchs JE, Huber MM, Bassi A, Lacetera A, Ruzsanyi V, Troppmair J, Amann A, Liedl KR. Precursors for cytochrome P450 profiling breath tests from an in silico screening approach. J Breath Res 2014; 8:046001. [PMID: 25233885 DOI: 10.1088/1752-7155/8/4/046001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The family of cytochrome P450 enzymes (CYPs) is a major player in the metabolism of drugs and xenobiotics. Genetic polymorphisms and transcriptional regulation give a complex patient-individual CYP activity profile for each human being. Therefore, personalized medicine demands easy and non-invasive measurement of the CYP phenotype. Breath tests detect volatile organic compounds (VOCs) in the patients' exhaled air after administration of a precursor molecule. CYP breath tests established for individual CYP isoforms are based on the detection of (13)CO2 or (14)CO2 originating from CYP-catalyzed oxidative degradation reactions of isotopically labeled precursors.We present an in silico work-flow aiming at the identification of novel precursor molecules, likely to result in VOCs other than CO2 upon oxidative degradation as we aim at label-free precursor molecules. The ligand-based work-flow comprises five parts: (1) CYP profiling was encoded as a decision tree based on 2D molecular descriptors derived from established models in the literature and validated against publicly available data extracted from the DrugBank. (2) Likely sites of metabolism were identified by reactivity and accessibility estimation for abstractable hydrogen radical. (3) Oxidative degradation reactions (O- and N-dealkylations) were found to be most promising in the release of VOCs. Thus, the CYP-catalyzed oxidative degradation reaction was encoded as SMIRKS (a programming language style to implement reactions based on the SMARTS description) to enumerate possible reaction products. (4) A quantitative structure property relation (QSPR) model aiming to predict the Henry constant H was derived from data for 488 organic compounds and identifies potentially VOCs amongst CYP reaction products. (5) A blacklist of naturally occurring breath components was implemented to identify marker molecules allowing straightforward detection within the exhaled air.Evident oxidative degradation reactions served as test case for the screening approach. Comparisons to metabolism data from literature support the results' plausibility. Thus, a large scale screening for potential novel breath test precursor using the presented five stage work-flow is promising.
Collapse
Affiliation(s)
- Susanne von Grafenstein
- Department of Theoretical Chemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innrain 80/82, A-6020 Innsbruck, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|