1
|
Paul JK, Malik A, Azmal M, Gulzar T, Afghan MTR, Talukder OF, Shahzadi S, Ghosh A. Advancing Alzheimer's Therapy: Computational strategies and treatment innovations. IBRO Neurosci Rep 2025; 18:270-282. [PMID: 39995567 PMCID: PMC11849200 DOI: 10.1016/j.ibneur.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 01/22/2025] [Accepted: 02/02/2025] [Indexed: 02/26/2025] Open
Abstract
Alzheimer's disease (AD) is a multifaceted neurodegenerative condition distinguished by the occurrence of memory impairment, cognitive deterioration, and neuronal impairment. Despite extensive research efforts, conventional treatment strategies primarily focus on symptom management, highlighting the need for innovative therapeutic approaches. This review explores the challenges of AD treatment and the integration of computational methodologies to advance therapeutic interventions. A comprehensive analysis of recent literature was conducted to elucidate the broad scope of Alzheimer's etiology and the limitations of conventional drug discovery approaches. Our findings underscore the critical role of computational models in elucidating disease mechanisms, identifying therapeutic targets, and expediting drug discovery. Through computational simulations, researchers can predict drug efficacy, optimize lead compounds, and facilitate personalized medicine approaches. Moreover, machine learning algorithms enhance early diagnosis and enable precision medicine strategies by analyzing multi-modal datasets. Case studies highlight the application of computational techniques in AD therapeutics, including the suppression of crucial proteins implicated in disease progression and the repurposing of existing drugs for AD management. Computational models elucidate the interplay between oxidative stress and neurodegeneration, offering insights into potential therapeutic interventions. Collaborative efforts between computational biologists, pharmacologists, and clinicians are essential to translate computational insights into clinically actionable interventions, ultimately improving patient outcomes and addressing the unmet medical needs of individuals affected by AD. Overall, integrating computational methodologies represents a promising paradigm shift in AD therapeutics, offering innovative solutions to overcome existing challenges and transform the landscape of AD treatment.
Collapse
Affiliation(s)
- Jibon Kumar Paul
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Abbeha Malik
- Department of Bioinformatics, Institute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, Pakistan
| | - Mahir Azmal
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Tooba Gulzar
- Department of Bioinformatics, Institute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, Pakistan
| | - Muhammad Talal Rahim Afghan
- Department of Bioinformatics, Institute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, Pakistan
| | - Omar Faruk Talukder
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Samar Shahzadi
- Department of Bioinformatics, Institute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, Pakistan
| | - Ajit Ghosh
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| |
Collapse
|
2
|
Karmakar S, Suman S, Uchoi J, Saha S, Dutta A, Das A, Kundu A. Essential Oils of Alpinia officinarum and Alpinia zerumbet: Potential Fungistatic Action and Molecular Dynamics Perspectives. Chem Biodivers 2025:e00221. [PMID: 40409754 DOI: 10.1002/cbdv.202500221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/25/2025]
Abstract
Valorisation of aromatic plants needs comprehensive research of bioactive phytochemicals through analytical and biochemical analysis. The present study focused on characterization of essential oils (EOs) of Alpinia officinarum (EOAO) and A. zerumbet (EOAZ) rhizomes for potential fungistatic action against selected decay-causing fungi, explaining their action through molecular dynamics (MD). The gas chromatography-mass spectrometry (GC-MS) analysis of EOAO and EOAZ revealed the identification of 35 and 53 compounds, representing 94.49% ± 3.10% and 96.46% ± 4.69% of the oil. Galangal acetate (46.91%) and 1,8-cineole (19.01%) were identified as the most abundant in EOAZ and EOAO and highly effective in arresting the growth of Penicillium expansum with EC50 14.74 and 21.02 µg mL-1, respectively. Fungistatic action followed the trend of P. expansum > Fusarium verticillioides > P. digitatum. Molecular docking and LigPlot+ analysis suggested the most favourable interactions of galangal acetate with the patulin synthase and aminotransferase, exhibiting docking scores of -8.68 kcal mol-1 (energy 41.79 kcal mol-1) and -6.25 kcal mol-1 (energy of -42.82 kcal mol-1), respectively. Galangal acetate was effective in blocking patulin synthase and aminotransferase, whereas terpinene-4-ol inhibited triacylglycerol lipase with multiple hydrogen bonds and π-π stacking, including strong hydrophobic and van der Waals interactions. Further, the MD of galangal acetate-patulin synthase complex revealed selective specificity and stability supported by values of deformability, B-factor, eigenvalues, residue index and variance. The presence of biologically active compounds makes Alpinia EOs a potential source of biofungicide, ensuring protection from fungal decay in agricultural produce.
Collapse
Affiliation(s)
- Subhrautpal Karmakar
- Division of Agricultural Chemicals, ICAR-Indian Agricultural Research Institute, New Delhi, India
- The Graduate School, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - Sourabh Suman
- Division of Agricultural Chemicals, ICAR-Indian Agricultural Research Institute, New Delhi, India
- The Graduate School, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - Julias Uchoi
- ICAR-National Bureau of Plant Genetic Resources, New Delhi, India
| | - Supradip Saha
- Division of Agricultural Chemicals, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - Anirban Dutta
- Downstream Agro-Processing Division, ICAR-National Institute of Secondary Agriculture, Ranchi, India
| | - Amrita Das
- Division of Plant Pathology, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - Aditi Kundu
- Division of Agricultural Chemicals, ICAR-Indian Agricultural Research Institute, New Delhi, India
| |
Collapse
|
3
|
Mondal S, Shrivastava P, Mehra R. Computing pathogenicity of mutations in human cytochrome P450 superfamily. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2025; 1873:141078. [PMID: 40349948 DOI: 10.1016/j.bbapap.2025.141078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/22/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Cytochrome P450 (CYPs) are crucial heme-containing enzymes that metabolize drugs and endogenous compounds. In humans, 57 CYP isoforms have been identified, with over 200 mutations linked to severe disorders. Our comprehensive computational study assessed the reason for the pathogenicity of mutations by comparing pathogenic and non-pathogenic variants. We analyzed 25,94,151 mutations across 26 CYP structures using structure- and sequence-based methods, revealing a meaningful stability pattern: non-pathogenic > all > pathogenic mutation datasets. Notably, pathogenic mutations were predominantly buried within CYP structures, indicating a higher potential for pathogenesis. We identified three key amino acid properties affected by mutations: Gibbs free energy, isoelectric point, and volume. Furthermore, diseased mutations significantly reduced positive residue content, particularly due to arginine mutations, which directly influenced the isoelectric point. Our findings indicate a greater likelihood of pathogenic mutations occurring at conserved sites, disrupting CYP function. A higher frequency of pathogenic mutations was observed in heme sites, primarily involving arginine, which may interfere with arginine-heme interactions. Molecular docking revealed a differential binding of heme in wild-type and pathogenic CYPs. This study provides a foundational analysis of mutation effects across multiple CYPs. It models the chemical basis of CYP-related pathogenicity, facilitating the development of a semi-quantitative disease prediction model.
Collapse
Affiliation(s)
- Somnath Mondal
- Department of Chemistry, Indian Institute of Technology Bhilai, Durg 491002, Chhattisgarh, India
| | - Pranchal Shrivastava
- Department of Chemistry, Indian Institute of Technology Bhilai, Durg 491002, Chhattisgarh, India
| | - Rukmankesh Mehra
- Department of Chemistry, Indian Institute of Technology Bhilai, Durg 491002, Chhattisgarh, India; Department of Bioscience and Biomedical Engineering, Indian Institute of Technology Bhilai, Durg 491002, Chhattisgarh, India.
| |
Collapse
|
4
|
Hu Y, Liu N, Ma C, Ren D, Wang D, Shang Y, Li F, Lyu Y, Cai C, Chen L, Liu W, Yu X. The Membrane-Targeting Synergistic Antifungal Effects of Walnut-Derived Peptide and Salicylic Acid on Prickly Pear Spoilage Fungus. Foods 2025; 14:951. [PMID: 40231962 PMCID: PMC11941157 DOI: 10.3390/foods14060951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/01/2025] [Accepted: 03/07/2025] [Indexed: 04/16/2025] Open
Abstract
Fermented walnut (FW) meal exhibits antifungal activity against Penicillium victoriae (the fungus responsible for prickly pear spoilage), which is mainly attributed to the synergistic effect of antimicrobial peptides and salicylic acid (SA). This study aimed to investigate the synergistic mechanism between YVVPW (YW-5, the peptide with the highest antifungal activity) and SA against the cell membrane of P. victoriae. Treatment enhanced prickly pear's rot rate, polyphenol concentration, and superoxide dismutase (SOD) activity by 38.11%, 8.11%, and 48.53%, respectively, while reducing the microbial count by 19.17%. Structural analyses revealed β-sheets as YW-5's predominant structure (41.18%), which increased to 49.0% during SA interaction. Molecular docking demonstrated YW-5's stronger binding to β-(1,3)-glucan synthase and membrane protein amino acids via hydrogen bonds, hydrophobic forces, and π-π conjugate interactions. Spectroscopic analyses demonstrated SA's major role in YW-5 synergy at the interface and polar head region of phospholipids, enhancing lipid chain disorder and the leakage of cell components. Malondialdehyde and SOD levels increased nearly two-fold and six-fold when treated with YW-5/SA, and YW-5 showed a more pronounced effect. Scanning electron and transmission electron microscopy confirmed that SA caused greater damage to spore morphology and cell ultrastructure. These findings support this formulation's functions as an efficient antifungal substance in fruit storage.
Collapse
Affiliation(s)
- Yue Hu
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China; (Y.H.); (N.L.); (C.M.); (D.W.); (Y.S.); (F.L.); (Y.L.); (C.C.); (L.C.); (W.L.)
| | - Na Liu
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China; (Y.H.); (N.L.); (C.M.); (D.W.); (Y.S.); (F.L.); (Y.L.); (C.C.); (L.C.); (W.L.)
| | - Caiqing Ma
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China; (Y.H.); (N.L.); (C.M.); (D.W.); (Y.S.); (F.L.); (Y.L.); (C.C.); (L.C.); (W.L.)
| | - Difeng Ren
- State Key Laboratory of Efficient Production of Forest Resources, Beijing Key Laboratory of Food Processing and Safety in Forestry, Department of Food Science and Engineering, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, China;
| | - Dujun Wang
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China; (Y.H.); (N.L.); (C.M.); (D.W.); (Y.S.); (F.L.); (Y.L.); (C.C.); (L.C.); (W.L.)
| | - Yueling Shang
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China; (Y.H.); (N.L.); (C.M.); (D.W.); (Y.S.); (F.L.); (Y.L.); (C.C.); (L.C.); (W.L.)
| | - Fengwei Li
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China; (Y.H.); (N.L.); (C.M.); (D.W.); (Y.S.); (F.L.); (Y.L.); (C.C.); (L.C.); (W.L.)
| | - Yongmei Lyu
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China; (Y.H.); (N.L.); (C.M.); (D.W.); (Y.S.); (F.L.); (Y.L.); (C.C.); (L.C.); (W.L.)
| | - Chen Cai
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China; (Y.H.); (N.L.); (C.M.); (D.W.); (Y.S.); (F.L.); (Y.L.); (C.C.); (L.C.); (W.L.)
| | - Long Chen
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China; (Y.H.); (N.L.); (C.M.); (D.W.); (Y.S.); (F.L.); (Y.L.); (C.C.); (L.C.); (W.L.)
| | - Wenjing Liu
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China; (Y.H.); (N.L.); (C.M.); (D.W.); (Y.S.); (F.L.); (Y.L.); (C.C.); (L.C.); (W.L.)
| | - Xiaohong Yu
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China; (Y.H.); (N.L.); (C.M.); (D.W.); (Y.S.); (F.L.); (Y.L.); (C.C.); (L.C.); (W.L.)
| |
Collapse
|
5
|
Biziukova NY, Rudik AV, Dmitriev AV, Tarasova OA, Filimonov DA, Poroikov VV. XenoMet: A Corpus of Texts to Extract Data on Metabolites of Xenobiotics. ACS OMEGA 2025; 10:2459-2471. [PMID: 39895765 PMCID: PMC11780559 DOI: 10.1021/acsomega.4c05723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/25/2024] [Accepted: 11/21/2024] [Indexed: 02/04/2025]
Abstract
Understanding the biotransformation of xenobiotics in the human body is critical for a comprehensive assessment of drug effects since pharmacologically active drug metabolites may exhibit a range of biological effects that often differ from those of the original pharmaceutical agent. Studies of the biotransformation mechanisms of xenobiotics have resulted in numerous publications. Extracting information about the parent compounds (substrates) and their metabolites from the texts allows retrieval of information on their biological activities, molecular mechanisms of action, and toxicity. Manual curation of the names of xenobiotics, their metabolites, and biotransformation reactions in the text is a challenging task due to the large number of publications related to studies of pharmaceutical agents metabolism. Our aim is to create an annotated corpus of texts that can be used for automated extraction of the names of xenobiotics, including pharmaceutical agents that undergo biotransformation and their metabolites. Prior to manual annotation of the corpus, semiautomatic annotation was carried out based on the earlier developed rule-based method for parent compounds and their metabolites extraction. To create XenoMet, we automatically extracted relevant texts from PubMed using a query based on MeSH terms. The names of biotransformation reactions were recognized by using an in-house-developed dictionary. Then, we manually verified the extracted data by correcting errors in the named entity annotation and identified the associations between substrates and metabolites. We tested the applicability of XenoMet for the reconstruction of a metabolic tree and for the automated extraction of the chemical names of substrates, metabolites, and reactions of biotransformation. Classification of the named entities of metabolites, substrates, and biotransformation reactions by a conditional random fields approach using XenoMet as the training set provides an F1-score of 0.79.
Collapse
Affiliation(s)
- Nadezhda Yu. Biziukova
- Institute of Biomedical
Chemistry, 10-8, Pogodinskaya
Str., Moscow 119121, Russian Federation
| | - Anastasia V. Rudik
- Institute of Biomedical
Chemistry, 10-8, Pogodinskaya
Str., Moscow 119121, Russian Federation
| | - Alexander V. Dmitriev
- Institute of Biomedical
Chemistry, 10-8, Pogodinskaya
Str., Moscow 119121, Russian Federation
| | - Olga A. Tarasova
- Institute of Biomedical
Chemistry, 10-8, Pogodinskaya
Str., Moscow 119121, Russian Federation
| | - Dmitry A. Filimonov
- Institute of Biomedical
Chemistry, 10-8, Pogodinskaya
Str., Moscow 119121, Russian Federation
| | - Vladimir V. Poroikov
- Institute of Biomedical
Chemistry, 10-8, Pogodinskaya
Str., Moscow 119121, Russian Federation
| |
Collapse
|
6
|
Zeng Z, Guo J, Jin J, Luo X. CLAIRE: a contrastive learning-based predictor for EC number of chemical reactions. J Cheminform 2025; 17:2. [PMID: 39773344 PMCID: PMC11707929 DOI: 10.1186/s13321-024-00944-8] [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: 07/23/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Predicting EC numbers for chemical reactions enables efficient enzymatic annotations for computer-aided synthesis planning. However, conventional machine learning approaches encounter challenges due to data scarcity and class imbalance. Here, we introduce CLAIRE (Contrastive Learning-based AnnotatIon for Reaction's EC), a novel framework leveraging contrastive learning, pre-trained language model-based reaction embeddings, and data augmentation to address these limitations. CLAIRE achieved notable performance improvements, demonstrating weighted average F1 scores of 0.861 and 0.911 on the testing set (n = 18,816) and an independent dataset (n = 1040) derived from yeast's metabolic model, respectively. Remarkably, CLAIRE significantly outperformed the state-of-the-art model by 3.65 folds and 1.18 folds, respectively. Its high accuracy positions CLAIRE as a promising tool for retrosynthesis planning, drug fate prediction, and synthetic biology applications. CLAIRE is freely available on GitHub ( https://github.com/zishuozeng/CLAIRE ).Scientific contributionThis work employed contrastive learning for predicting enzymatic reaction's EC numbers, overcoming the challenges in data scarcity and imbalance. The new model achieves the state-of-the-art performance and may facilitate the computer-aided synthesis planning.
Collapse
Affiliation(s)
- Zishuo Zeng
- Synceres Biosciences Co. Ltd., Shenzhen, 518100, China.
| | - Jin Guo
- Synceres Biosciences Co. Ltd., Shenzhen, 518100, China
| | - Jiao Jin
- Synceres Biosciences Co. Ltd., Shenzhen, 518100, China
| | - Xiaozhou Luo
- Shenzhen Key Laboratory for the Intelligent Microbial Manufacturing of Medicines, Key Laboratory of Quantitative Synthetic Biology, Center for Synthetic Biochemistry, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
7
|
Wei Y, Palazzolo L, Ben Mariem O, Bianchi D, Laurenzi T, Guerrini U, Eberini I. Investigation of in silico studies for cytochrome P450 isoforms specificity. Comput Struct Biotechnol J 2024; 23:3090-3103. [PMID: 39188968 PMCID: PMC11347072 DOI: 10.1016/j.csbj.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 08/28/2024] Open
Abstract
Cytochrome P450 (CYP450) enzymes comprise a highly diverse superfamily of heme-thiolate proteins that responsible for catalyzing over 90 % of enzymatic reactions associated with xenobiotic metabolism in humans. Accurately predicting whether chemicals are substrates or inhibitors of different CYP450 isoforms can aid in pre-selecting hit compounds for the drug discovery process, chemical toxicology studies, and patients treatment planning. In this work, we investigated in silico studies on CYP450s specificity over past twenty years, categorizing these studies into structure-based and ligand-based approaches. Subsequently, we utilized 100 of the most frequently prescribed drugs to test eleven machine learning-based prediction models which were published between 2015 and 2024. We analyzed various aspects of the evaluated models, such as their datasets, algorithms, and performance. This will give readers with a comprehensive overview of these prediction models and help them choose the most suitable one to do prediction. We also provide our insights for future research trend in both structure-based and ligand-based approaches in this field.
Collapse
Affiliation(s)
- Yao Wei
- Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti”, Università degli Studi di Milano, Via Giuseppe Balzaretti 9, 20133 Milano, Italy
| | - Luca Palazzolo
- Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti”, Università degli Studi di Milano, Via Giuseppe Balzaretti 9, 20133 Milano, Italy
| | - Omar Ben Mariem
- Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti”, Università degli Studi di Milano, Via Giuseppe Balzaretti 9, 20133 Milano, Italy
| | - Davide Bianchi
- Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti”, Università degli Studi di Milano, Via Giuseppe Balzaretti 9, 20133 Milano, Italy
| | - Tommaso Laurenzi
- Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti”, Università degli Studi di Milano, Via Giuseppe Balzaretti 9, 20133 Milano, Italy
| | - Uliano Guerrini
- Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti”, Università degli Studi di Milano, Via Giuseppe Balzaretti 9, 20133 Milano, Italy
| | - Ivano Eberini
- Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti”, Università degli Studi di Milano, Via Giuseppe Balzaretti 9, 20133 Milano, Italy
| |
Collapse
|
8
|
Janssen K, Kirchmair J, Proppe J. Relevance and Potential Applications of C2-Carboxylated 1,3-Azoles. ChemMedChem 2024; 19:e202400307. [PMID: 39022854 DOI: 10.1002/cmdc.202400307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/20/2024]
Abstract
Carbon dioxide (CO2) is an economically viable and abundant carbon source that can be incorporated into compounds such as C2-carboxylated 1,3-azoles relevant to the pharmaceutical, cosmetics, and pesticide industries. Of the 2.4 million commercially available C2-unsubstituted 1,3-azole compounds, less than 1 % are currently purchasable as their C2-carboxylated derivatives, highlighting the substantial gap in compound availability. This availability gap leaves ample opportunities for exploring the synthetic accessibility and use of carboxylated azoles in bioactive compounds. In this study, we analyze and quantify the relevance of C2-carboxylated 1,3-azoles in small-molecule research. An analysis of molecular databases such as ZINC, ChEMBL, COSMOS, and DrugBank identified relevant C2-carboxylated 1,3-azoles as anticoagulant and aroma-giving compounds. Moreover, a pharmacophore analysis highlights promising pharmaceutical potential associated with C2-carboxylated 1,3-azoles, revealing the ATP-sensitive inward rectifier potassium channel 1 (KATP) and Kinesin-like protein KIF18 A as targets that can potentially be addressed with C2-carboxylated 1,3-azoles. Moreover, we identified several bioisosteres of C2-carboxylated 1,3-azoles. In conclusion, further exploration of the chemical space of C2-carboxylated 1,3-azoles is recommended to harness their full potential in drug discovery and related fields.
Collapse
Affiliation(s)
- Kerrin Janssen
- Institute of Physical and Theoretical Chemistry, TU Braunschweig, 38106, Braunschweig, Germany
| | - Johannes Kirchmair
- Christian Doppler Laboratory for Molecular Informatics in the Biosciences and Department of Pharmaceutical Sciences, University of Vienna, 1090, Vienna, Austria
| | - Jonny Proppe
- Institute of Physical and Theoretical Chemistry, TU Braunschweig, 38106, Braunschweig, Germany
| |
Collapse
|
9
|
Vyas SK, Das A, Suryanarayana Murty U, Dixit VA. Sulfotransferase-mediated phase II drug metabolism prediction of substrates and sites using accessibility and reactivity-based algorithms. Mol Inform 2024; 43:e202400008. [PMID: 39110066 DOI: 10.1002/minf.202400008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 05/18/2024] [Accepted: 06/24/2024] [Indexed: 10/16/2024]
Abstract
Sulphotransferases (SULTs) are a major phase II metabolic enzyme class contributing ~20 % to the Phase II metabolism of FDA-approved drugs. Ignoring the potential for SULT-mediated metabolism leaves a strong potential for drug-drug interactions, often causing late-stage drug discovery failures or black-boxed warnings on FDA labels. The existing models use only accessibility descriptors and machine learning (ML) methods for class and site of sulfonation (SOS) predictions for SULT. In this study, a variety of accessibility, reactivity, and hybrid models and algorithms have been developed to make accurate substrate and SOS predictions. Unlike the literature models, reactivity parameters for the aliphatic or aromatic hydroxyl groups (R/Ar-O-H), the Bond Dissociation Energy (BDE) gave accurate models with a True Positive Rate (TPR)=0.84 for SOS predictions. We offer mechanistic insights to explain these novel findings that are not recognized in the literature. The accessibility parameters like the ratio of Chemgauss4 Score (CGS) and Molecular Weight (MW) CGS/MW and distance from cofactor (Dis) were essential for class predictions and showed TPR=0.72. Substrates consistently had lower BDE, Dis, and CGS/MW than non-substrates. Hybrid models also performed acceptablely for SOS predictions. Using the best models, Algorithms gave an acceptable performance in class prediction: TPR=0.62, False Positive Rate (FPR)=0.24, Balanced accuracy (BA)=0.69, and SOS prediction: TPR=0.98, FPR=0.60, and BA=0.69. A rule-based method was added to improve the predictive performance, which improved the algorithm TPR, FPR, and BA. Validation using an external dataset of drug-like compounds gave class prediction: TPR=0.67, FPR=0.00, and SOS prediction: TPR=0.80 and FPR=0.44 for the best Algorithm. Comparisons with standard ML models also show that our algorithm shows higher predictive performance for classification on external datasets. Overall, these models and algorithms (SOS predictor) give accurate substrate class and site (SOS) predictions for SULT-mediated Phase II metabolism and will be valuable to the drug discovery community in academia and industry. The SOS predictor is freely available for academic/non-profit research via the GitHub link.
Collapse
Affiliation(s)
- Shivam Kumar Vyas
- Department of Medicinal Chemistry, Department of Pharmaceuticals, Ministry of Chemicals & Fertilizers, Govt. of India, Sila Katamur (Halugurisuk), P.O.: Changsari, Dist: Kamrup, Pin, National Institute of Pharmaceutical Education and Research, Guwahati, (NIPER Guwahati), Guwahati, Assam, 781101, India
| | - Avik Das
- Department of Pharmacy, Birla Institute of Technology and Sciences Pilani (BITS-Pilani), Vidya Vihar Campus 41, Pilani, Rajasthan, 333031, India
- Current address: Department of Primary Intelligence, IQVIA, Sarjapur-Marathahalli Outer Ring Road Embassy Tech Square, Bangalore, 560103 Karnataka, India
| | - Upadhyayula Suryanarayana Murty
- Department of Medicinal Chemistry, Department of Pharmaceuticals, Ministry of Chemicals & Fertilizers, Govt. of India, Sila Katamur (Halugurisuk), P.O.: Changsari, Dist: Kamrup, Pin, National Institute of Pharmaceutical Education and Research, Guwahati, (NIPER Guwahati), Guwahati, Assam, 781101, India
| | - Vaibhav A Dixit
- Department of Medicinal Chemistry, Department of Pharmaceuticals, Ministry of Chemicals & Fertilizers, Govt. of India, Sila Katamur (Halugurisuk), P.O.: Changsari, Dist: Kamrup, Pin, National Institute of Pharmaceutical Education and Research, Guwahati, (NIPER Guwahati), Guwahati, Assam, 781101, India
| |
Collapse
|
10
|
Guvench O. Effect of Lipid Bilayer Anchoring on the Conformational Properties of the Cytochrome P450 2D6 Binding Site. J Phys Chem B 2024; 128:7188-7198. [PMID: 39016537 DOI: 10.1021/acs.jpcb.4c03097] [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: 07/18/2024]
Abstract
Human cytochrome P450 (CYP) proteins metabolize 75% of small-molecule pharmaceuticals, which makes structure-based modeling of CYP metabolism and inhibition, bolstered by the current availability of X-ray crystal structures of CYP globular catalytic domains, an attractive prospect. Accounting for this broad metabolic capacity is a combination of the existence of multiple different CYP proteins and the capacity of a single CYP protein to metabolize multiple different small molecules. It is thought that structural plasticity and flexibility contribute to this latter property; therefore, incorporating diverse conformational states of a particular CYP is likely an important consideration in structure-based CYP metabolism and inhibition modeling. While all-atom explicit-solvent molecular dynamics simulations can be used to generate conformational ensembles under biologically relevant conditions, existing CYP crystal structures are of the globular domain only, whereas human CYPs contain N-terminal transmembrane and linker peptides that anchor the globular catalytic domain to the endoplasmic reticulum. To determine whether this can cause significant differences in the sampled binding site conformations, microsecond scale all-atom explicit-solvent molecular dynamics simulations of the CYP2D6 globular domain in an aqueous environment were compared with those of the full-length protein anchored in a POPC lipid bilayer. While bilayer-anchoring damped some structural fluctuations in the globular domain relative to the aqueous simulations, none of the affected residues included binding site pocket residues. Furthermore, clustering of molecular dynamics snapshots based on either pairwise binding site pocket RMSD or volume differences demonstrated a lack of separation of snapshots from the two simulation conditions into different clusters. These results suggest the substantially simpler and computationally cheaper aqueous simulation approach can be used to generate a relevant conformational ensemble of the CYP2D6 binding site for structure-based metabolism and inhibition modeling.
Collapse
Affiliation(s)
- Olgun Guvench
- Department of Pharmaceutical Sciences and Administration, School of Pharmacy, Westbrook College of Health Professions, University of New England, 716 Stevens Ave, Portland, Maine 04103, United States
| |
Collapse
|
11
|
Smith DA, Burton LM, Smith SA. Through a computer monitor darkly: artificial intelligence in absorption, distribution, metabolism and excretion science. Xenobiotica 2024; 54:359-367. [PMID: 38095217 DOI: 10.1080/00498254.2023.2295361] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/12/2023] [Indexed: 08/22/2024]
Abstract
Artificial Intelligence (AI) is poised or has already begun to influence absorption, distribution, metabolism and excretion (ADME) science. It is not in the area expected - that of superior modelling of ADME data to increase its predictive power. It is influencing traditional exhaustive and careful literature research by providing almost perfect summaries of existing information. This will highly influence how people study, graduate and progress in the ADME sciences. The literature contains many flaws (protein binding influence on unbound drug concentration is one of the examples cited) and without direction AI may help to popularise them.ADME science has a relatively small number of key assays and values, but these are produced under widely varying conditions so large data sets, the best substrate for artificial intelligence, are not readily available to produce new more predictive systems. The use of AI to enrich the databases may be a near term goal.AI is already contributing in other areas such as technical skill assimilation, maintenance of complex instruments (combined with virtual reality) and the processing of pharmacovigilance.
Collapse
|
12
|
Tang W, Shen T, Chen Z. In silico discovery of potential PPI inhibitors for anti-lung cancer activity by targeting the CCND1-CDK4 complex via the P21 inhibition mechanism. Front Chem 2024; 12:1404573. [PMID: 38957406 PMCID: PMC11217521 DOI: 10.3389/fchem.2024.1404573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/31/2024] [Indexed: 07/04/2024] Open
Abstract
Non-Small Cell Lung Cancer (NSCLC) is a prevalent and deadly form of lung cancer worldwide with a low 5-year survival rate. Current treatments have limitations, particularly for advanced-stage patients. P21, a protein that inhibits the CCND1-CDK4 complex, plays a crucial role in cell proliferation. Computer-Aided Drug Design (CADD) based on pharmacophores can screen and design PPI inhibitors targeting the CCND1-CDK4 complex. By analyzing known inhibitors, key pharmacophores are identified, and computational methods are used to screen potential PPI inhibitors. Molecular docking, pharmacophore matching, and structure-activity relationship studies optimize the inhibitors. This approach accelerates the discovery of CCND1-CDK4 PPI inhibitors for NSCLC treatment. Molecular dynamics simulations of CCND1-CDK4-P21 and CCND1-CDK4 complexes showed stable behavior, comprehensive sampling, and P21's impact on complex stability and hydrogen bond formation. A pharmacophore model facilitated virtual screening, identifying compounds with favorable binding affinities. Further simulations confirmed the stability and interactions of selected compounds, including 513457. This study demonstrates the potential of CADD in optimizing PPI inhibitors targeting the CCND1-CDK4 complex for NSCLC treatment. Extended simulations and experimental validations are necessary to assess their efficacy and safety.
Collapse
Affiliation(s)
| | | | - Zhoumiao Chen
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
13
|
Groff L, Williams A, Shah I, Patlewicz G. MetSim: Integrated Programmatic Access and Pathway Management for Xenobiotic Metabolism Simulators. Chem Res Toxicol 2024; 37:685-697. [PMID: 38598715 PMCID: PMC11325951 DOI: 10.1021/acs.chemrestox.3c00398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Xenobiotic metabolism is a key consideration in evaluating the hazards and risks posed by environmental chemicals. A number of software tools exist that are capable of simulating metabolites, but each reports its predictions in a different format and with varying levels of detail. This makes comparing the performance and coverage of the tools a practical challenge. To address this shortcoming, we developed a metabolic simulation framework called MetSim, which comprises three main components. A graph-based schema was developed to allow metabolism information to be harmonized. The schema was implemented in MongoDB to store and retrieve metabolic graphs for subsequent analysis. MetSim currently includes an application programming interface for four metabolic simulators: BioTransformer, the OECD Toolbox, EPA's chemical transformation simulator (CTS), and tissue metabolism simulator (TIMES). Lastly, MetSim provides functions to help evaluate simulator performance for specific data sets. In this study, a set of 112 drugs with 432 reported metabolites were compiled, and predictions were made using the 4 simulators. Fifty-nine of the 112 drugs were taken from the Small Molecule Pathway Database, with the remainder sourced from the literature. The human models within BioTransformer and CTS (Phase I only) and the rat models within TIMES and the OECD Toolbox (Phase I only) were used to make predictions for the chemicals in the data set. The recall and precision (recall, precision) ranked in order of highest recall for each individual tool were CTS (0.54, 0.017), BioTransformer (0.50, 0.008), Toolbox in vitro (0.40, 0.144), TIMES in vivo (0.40, 0.133), Toolbox in vivo (0.40, 0.118), and TIMES in vitro (0.39, 0.128). Combining all of the model predictions together increased the overall recall (0.73, 0.008). MetSim enabled insights into the performance and coverage of in silico metabolic simulators to be more efficiently derived, which in turn should aid future efforts to evaluate other data sets.
Collapse
Affiliation(s)
- Louis Groff
- Center for Computational Toxicology and Exposure (CCTE), Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Antony Williams
- Center for Computational Toxicology and Exposure (CCTE), Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Imran Shah
- Center for Computational Toxicology and Exposure (CCTE), Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Grace Patlewicz
- Center for Computational Toxicology and Exposure (CCTE), Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| |
Collapse
|
14
|
Hu Y, Ling Y, Qin Z, Huang J, Jian L, Ren DF. Isolation, identification, and synergistic mechanism of a novel antimicrobial peptide and phenolic compound from fermented walnut meal and their application in Rosa roxbughii Tratt spoilage fungus. Food Chem 2024; 433:137333. [PMID: 37696092 DOI: 10.1016/j.foodchem.2023.137333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 04/26/2023] [Accepted: 08/28/2023] [Indexed: 09/13/2023]
Abstract
This study aimed to identify an antimicrobial peptide and phenolic compound combination derived from fermented walnut meal against Penicillium. victoriae, a fungus responsible for Rosa. roxbughii Tratt spoilage, and ultimately investigate their synergistic mechanism. YVVPW and salicylic acid (SA) had the highest antifungal activity among identified 4 antimicrobial peptides, including FGGDSTHP, ALGGGY, YVVPW, and PLLRW, and 15 phenolic compounds, respectively. Molecular docking verified that YVVPW bound to regulatory subunit via hydrogen-bond, hydrophobic, and π-π conjugate interactions. YVVPW and SA exhibited synergistic effects with average minimal inhibitory concentration decreasing by 85.44 ± 8.04%. Fluorescence spectroscopy demonstrated quenching of intrinsic Trp and Tyr fluorescence by interaction. FTIR and molecular docking results revealed formation of 3 hydrogen bonds via OH, CO, NH, and CH bonds in YVVPW + SA, with π-π stacking occurring between the benzene ring and five-membered ring. These reinforce potential application of this combination as an effective fungistatic combination in fruit preservation.
Collapse
Affiliation(s)
- Yue Hu
- State Key Laboratory of Efficient Production of Forest Resources, Beijing Key Laboratory of Food Processing and Safety in Forestry, Department of Food Science and Engineering, College of Biological Sciences and Biotechnology, Beijing Forestry University, 100083 Beijing, China.
| | - Yuxi Ling
- State Key Laboratory of Efficient Production of Forest Resources, Beijing Key Laboratory of Food Processing and Safety in Forestry, Department of Food Science and Engineering, College of Biological Sciences and Biotechnology, Beijing Forestry University, 100083 Beijing, China.
| | - Zhouyi Qin
- State Key Laboratory of Efficient Production of Forest Resources, Beijing Key Laboratory of Food Processing and Safety in Forestry, Department of Food Science and Engineering, College of Biological Sciences and Biotechnology, Beijing Forestry University, 100083 Beijing, China.
| | - Jingmei Huang
- State Key Laboratory of Efficient Production of Forest Resources, Beijing Key Laboratory of Food Processing and Safety in Forestry, Department of Food Science and Engineering, College of Biological Sciences and Biotechnology, Beijing Forestry University, 100083 Beijing, China.
| | - Liuyu Jian
- State Key Laboratory of Efficient Production of Forest Resources, Beijing Key Laboratory of Food Processing and Safety in Forestry, Department of Food Science and Engineering, College of Biological Sciences and Biotechnology, Beijing Forestry University, 100083 Beijing, China.
| | - Di Feng Ren
- State Key Laboratory of Efficient Production of Forest Resources, Beijing Key Laboratory of Food Processing and Safety in Forestry, Department of Food Science and Engineering, College of Biological Sciences and Biotechnology, Beijing Forestry University, 100083 Beijing, China.
| |
Collapse
|
15
|
Dai Z, Wu Y, Xiong Y, Wu J, Wang M, Sun X, Ding X, Yang L, Sun X, Ge G. CYP1A inhibitors: Recent progress, current challenges, and future perspectives. Med Res Rev 2024; 44:169-234. [PMID: 37337403 DOI: 10.1002/med.21982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/28/2023] [Accepted: 05/23/2023] [Indexed: 06/21/2023]
Abstract
Mammalian cytochrome P450 1A (CYP1A) are key phase I xenobiotic-metabolizing enzymes that play a distinctive role in metabolic activation or metabolic clearance of a variety of procarcinogens, drugs, and endogenous substances. Human CYP1A subfamily contains two members (hCYP1A1 and hCYP1A2), which are known to catalyze the oxidative activation of some environmental procarcinogens into carcinogenic species. Increasing evidence has demonstrated that CYP1A inhibitor therapies are promising strategies for cancer chemoprevention or overcoming CYP1A-associated drug toxicity and resistance. Herein, we reviewed recent advances in the discovery and characterization of hCYP1A inhibitors, from the discovery approaches to structural features and biomedical applications of hCYP1A inhibitors. The inhibition potentials, inhibition modes, and inhibition constants of all reported hCYP1A inhibitors are comprehensively summarized. Meanwhile, the structural features and structure-activity relationships of different classes of hCYP1A1 and hCYP1A2 inhibitors are analyzed and discussed in depth. Furthermore, the major challenges and future directions for this field are presented and highlighted. Collectively, the information and knowledge presented here will strongly facilitate the researchers to discover and develop more efficacious CYP1A inhibitors for specific purposes, such as chemo-preventive agents or as tool molecules in hCYP1A-related fundamental studies.
Collapse
Affiliation(s)
- Ziru Dai
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yue Wu
- Shanghai Frontiers Science Center for TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuan Xiong
- Shanghai Frontiers Science Center for TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingjing Wu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Min Wang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiao Sun
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xinxin Ding
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, America
| | - Ling Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Xiaobo Sun
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Guangbo Ge
- Shanghai Frontiers Science Center for TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
16
|
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
|
17
|
Trostel L, Coll C, Fenner K, Hafner J. Combining predictive and analytical methods to elucidate pharmaceutical biotransformation in activated sludge. ENVIRONMENTAL SCIENCE. PROCESSES & IMPACTS 2023; 25:1322-1336. [PMID: 37539453 DOI: 10.1039/d3em00161j] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
While man-made chemicals in the environment are ubiquitous and a potential threat to human health and ecosystem integrity, the environmental fate of chemical contaminants such as pharmaceuticals is often poorly understood. Biodegradation processes driven by microbial communities convert chemicals into transformation products (TPs) that may themselves have adverse ecological effects. The detection of TPs formed during biodegradation has been continuously improved thanks to the development of TP prediction algorithms and analytical workflows. Here, we contribute to this advance by (i) reviewing past applications of TP identification workflows, (ii) applying an updated workflow for TP prediction to 42 pharmaceuticals in biodegradation experiments with activated sludge, and (iii) benchmarking 5 different pathway prediction models, comprising 4 prediction models trained on different datasets provided by enviPath, and the state-of-the-art EAWAG pathway prediction system. Using the updated workflow, we could tentatively identify 79 transformation products for 31 pharmaceutical compounds. Compared to previous works, we have further automatized several steps that were previously performed by hand. By benchmarking the enviPath prediction system on experimental data, we demonstrate the usefulness of the pathway prediction tool to generate suspect lists for screening, and we propose new avenues to improve their accuracy. Moreover, we provide a well-documented workflow that can be (i) readily applied to detect transformation products in activated sludge and (ii) potentially extended to other environmental studies.
Collapse
Affiliation(s)
- Leo Trostel
- Department of Environmental Chemistry, Swiss Federal Institute of Aquatic Science and Technology (Eawag), Dübendorf, 8600, Zürich, Switzerland.
| | - Claudia Coll
- Department of Environmental Chemistry, Swiss Federal Institute of Aquatic Science and Technology (Eawag), Dübendorf, 8600, Zürich, Switzerland.
| | - Kathrin Fenner
- Department of Environmental Chemistry, Swiss Federal Institute of Aquatic Science and Technology (Eawag), Dübendorf, 8600, Zürich, Switzerland.
- Department of Chemistry, University of Zürich, 8057 Zürich, Switzerland
| | - Jasmin Hafner
- Department of Environmental Chemistry, Swiss Federal Institute of Aquatic Science and Technology (Eawag), Dübendorf, 8600, Zürich, Switzerland.
- Department of Chemistry, University of Zürich, 8057 Zürich, Switzerland
| |
Collapse
|
18
|
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
|
19
|
Azad I, Khan T, Ahmad N, Khan AR, Akhter Y. Updates on drug designing approach through computational strategies: a review. Future Sci OA 2023; 9:FSO862. [PMID: 37180609 PMCID: PMC10167725 DOI: 10.2144/fsoa-2022-0085] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/12/2023] [Indexed: 05/16/2023] Open
Abstract
The drug discovery and development (DDD) process in pursuit of novel drug candidates is a challenging procedure requiring lots of time and resources. Therefore, computer-aided drug design (CADD) methodologies are used extensively to promote proficiency in drug development in a systematic and time-effective manner. The point in reference is SARS-CoV-2 which has emerged as a global pandemic. In the absence of any confirmed drug moiety to treat the infection, the science fraternity adopted hit and trial methods to come up with a lead drug compound. This article is an overview of the virtual methodologies, which assist in finding novel hits and help in the progression of drug development in a short period with a specific medicinal solution.
Collapse
Affiliation(s)
- Iqbal Azad
- Department of Chemistry, Integral University, Dasauli, P.O. Bas-ha, Kursi Road, Lucknow, 226026, UP, India
| | - Tahmeena Khan
- Department of Chemistry, Integral University, Dasauli, P.O. Bas-ha, Kursi Road, Lucknow, 226026, UP, India
| | - Naseem Ahmad
- Department of Chemistry, Integral University, Dasauli, P.O. Bas-ha, Kursi Road, Lucknow, 226026, UP, India
| | - Abdul Rahman Khan
- Department of Chemistry, Integral University, Dasauli, P.O. Bas-ha, Kursi Road, Lucknow, 226026, UP, India
| | - Yusuf Akhter
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, UP, 2260025, India
| |
Collapse
|
20
|
Ota R, Yamashita F. Application of machine learning techniques to the analysis and prediction of drug pharmacokinetics. J Control Release 2022; 352:961-969. [PMID: 36370876 DOI: 10.1016/j.jconrel.2022.11.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 10/23/2022] [Accepted: 11/07/2022] [Indexed: 11/17/2022]
Abstract
In this review, we describe the current status and challenges in applying machine-learning techniques to the analysis and prediction of pharmacokinetic data. The theory of pharmacokinetics has been developed over decades on the basis of physiology and reaction kinetics. Mathematical models allow the reduction of pharmacokinetic data to parameter values, giving insight and understanding into ADME processes and predicting the outcome of different dosing scenarios. However, much information hidden in the data is lost through conceptual simplification with models. It is difficult to use mechanistic models alone to predict diverse pharmacokinetic time profiles, including inter-drug and inter-individual differences, in a cross-sectional manner. Machine learning is a prediction platform that can handle complex phenomena through data-driven analysis. As a resule, machine learning has been successfully adopted in various fields, including image recognition and language processing, and has been used for over two decades in pharmacokinetic research, primarily in the area of quantitative structure-activity relationships for pharmacokinetic parameters. Machine-learning models are generally known to provide better predictive performance than conventional linear models. Owing to the recent success in deep learning, models with new structures are being consistently proposed. These models include transfer learning and generative adversarial networks, which contribute to the effective use of a limited amount of data by diverting existing similar models or generating pseudo-data. How to make such newly emerging machine learning technologies applicable to meet challenges in the pharmacokinetics/pharmacodynamics field is now the key issue.
Collapse
Affiliation(s)
- Ryosaku Ota
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Fumiyoshi Yamashita
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan; Department of Applied Pharmacy and Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
21
|
Rzepiela AA, Viarengo-Baker LA, Tatarskii V, Kombarov R, Whitty A. Conformational Effects on the Passive Membrane Permeability of Synthetic Macrocycles. J Med Chem 2022; 65:10300-10317. [PMID: 35861996 DOI: 10.1021/acs.jmedchem.1c02090] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Macrocyclic compounds (MCs) can have complex conformational properties that affect pharmacologically important behaviors such as membrane permeability. We measured the passive permeability of 3600 diverse nonpeptidic MCs and used machine learning to analyze the results. Incorporating selected properties based on the three-dimensional (3D) conformation gave models that predicted permeability with Q2 = 0.81. A biased spatial distribution of polar versus nonpolar regions was particularly important for good permeability, consistent with a mechanism in which the initial insertion of nonpolar portions of a MC helps facilitate the subsequent membrane entry of more polar parts. We also examined effects on permeability of 800 substructural elements by comparing matched molecular pairs. Some substitutions were invariably beneficial or invariably deleterious to permeability, while the influence of others was highly contextual. Overall, the work provides insights into how the permeability of MCs is influenced by their 3D conformational properties and suggests design hypotheses for achieving macrocycles with high membrane permeability.
Collapse
Affiliation(s)
- Anna A Rzepiela
- Pyxis Discovery, Delftechpark 26, 2628XH Delft, The Netherlands
| | - Lauren A Viarengo-Baker
- Department of Chemistry, Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States
| | - Victor Tatarskii
- Asinex Corporation, 101 N Chestnut St # 104, Winston-Salem, North Carolina 27101,United States
| | - Roman Kombarov
- Asinex Corporation, 101 N Chestnut St # 104, Winston-Salem, North Carolina 27101,United States
| | - Adrian Whitty
- Department of Chemistry, Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States.,Center for Molecular Discovery, Boston University, 24 Cummington Mall, Boston, Massachusetts 02215, United States
| |
Collapse
|
22
|
Smith AME, Lanevskij K, Sazonovas A, Harris J. Impact of Established and Emerging Software Tools on the Metabolite Identification Landscape. FRONTIERS IN TOXICOLOGY 2022; 4:932445. [PMID: 35800176 PMCID: PMC9253584 DOI: 10.3389/ftox.2022.932445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 05/30/2022] [Indexed: 11/25/2022] Open
Abstract
Scientists' ability to detect drug-related metabolites at trace concentrations has improved over recent decades. High-resolution instruments enable collection of large amounts of raw experimental data. In fact, the quantity of data produced has become a challenge due to effort required to convert raw data into useful insights. Various cheminformatics tools have been developed to address these metabolite identification challenges. This article describes the current state of these tools. They can be split into two categories: Pre-experimental metabolite generation and post-experimental data analysis. The former can be subdivided into rule-based, machine learning-based, and docking-based approaches. Post-experimental tools help scientists automatically perform chromatographic deconvolution of LC/MS data and identify metabolites. They can use pre-experimental predictions to improve metabolite identification, but they are not limited to these predictions: unexpected metabolites can also be discovered through fractional mass filtering. In addition to a review of available software tools, we present a description of pre-experimental and post-experimental metabolite structure generation using MetaSense. These software tools improve upon manual techniques, increasing scientist productivity and enabling efficient handling of large datasets. However, the trend of increasingly large datasets and highly data-driven workflows requires a more sophisticated informatics transition in metabolite identification labs. Experimental work has traditionally been separated from the information technology tools that handle our data. We argue that these IT tools can help scientists draw connections via data visualizations and preserve and share results via searchable centralized databases. In addition, data marshalling and homogenization techniques enable future data mining and machine learning.
Collapse
|
23
|
Metazoan stringent-like response mediated by MESH1 phenotypic conservation via distinct mechanisms. Comput Struct Biotechnol J 2022; 20:2680-2684. [PMID: 35685369 PMCID: PMC9166373 DOI: 10.1016/j.csbj.2022.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/30/2022] [Accepted: 05/01/2022] [Indexed: 12/27/2022] Open
Abstract
All organisms are constantly exposed to various stresses, necessitating adaptive strategies for survival. In bacteria, the main metabolic stress-coping mechanism is the stringent response, which is triggered by the accumulation of “alarmone” (p)ppGpp to arrest proliferation and reprogram the transcriptome. The level of (p)ppGpp is regulated by its synthetase RelA and its hydrolase SpoT. MESH1 is the metazoan homolog of bacterial SpoT that regulates the bacterial stringent response by degrading the alarmone (p)ppGpp. While MESH1, like SpoT, can also dephosphorylate (p)ppGpp, mammalian cells do not have significant levels of this metabolite, and the relevant enzymatic activities and function of MESH1 have remained a mystery. Through genetic and biochemical analyses, we have solved the long-held mystery and identified MESH1 as the first mammalian cytosolic NADPH phosphatase involved in ferroptosis. Furthermore, we discovered that MESH1 removal leads to proliferation arrest, translation inhibition, and a prominent transcriptional and metabolic response. Therefore, MESH1 knockdown triggers a novel stress response with phenotypic conservation with the bacterial stringent response via distinct substrates and molecular pathways. Here, we summarize the background of the MESH1, illustrate the striking conservation of phenotypes in different organisms during evolution and discuss remaining questions in the field.
Collapse
|
24
|
Zheng S, Wang L, Xiong J, Liang G, Xu Y, Lin F. Consensus Prediction of Human Gut Microbiota-Mediated Metabolism Susceptibility for Small Molecules by Machine Learning, Structural Alerts, and Dietary Compounds-Based Average Similarity Methods. J Chem Inf Model 2022; 62:1078-1099. [PMID: 35156807 DOI: 10.1021/acs.jcim.1c00948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The human gut microbiota (HGM) colonizing human gastrointestinal tract (HGT) confers a repertoire of dynamic and unique metabolic capacities that are not possessed by the host and therefore is tentatively perceived as an alternative metabolic ″organ″ besides the liver in the host. Nevertheless, the significant contribution of HGM to the overall human metabolism is often overlooked in the modern drug discovery pipeline. Hence, a systematic evaluation of HGM-mediated drug metabolism is gradually important, and its computational prediction becomes increasingly necessary. In this work, a new data set containing both the HGM-mediated metabolism susceptible (HGMMS) and insusceptible (HGMMI) compounds (329 vs 320) was manually curated. Based on this data set, the first machine learning (ML) model, a new structural alerts (SA) model, and the K-nearest neighboring dietary compounds-based average similarity (AS) model were proposed to directly predict the HGM-mediated metabolism susceptibility for small molecules, and exhibit promising performance on three independent test sets. Finally, consensus prediction (ML/SA/AS) for DrugBank molecules revealed an intriguing phenomenon that a typical Michael acceptor ″α,β-unsaturated carbonyl group″ is a very common warhead for the design of covalent inhibitors and inclined to be metabolized by HGM in anaerobic HGT to generate the reduced metabolite without the reactive warhead, which could be a new concern to medicinal chemists. To the best of our knowledge, we gleaned the first HGMMS/HGMMI data set, developed the first HGMMS/HGMMI classification model, implemented a relatively comprehensive program based on ML/SA/AS approaches, and found a new phenomenon on the HGM-mediated deactivation of an extensively used warhead for covalent inhibitors.
Collapse
Affiliation(s)
- Suqing Zheng
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China.,Chemical Biology Research Center, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Lei Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Jun Xiong
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Guang Liang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China.,Chemical Biology Research Center, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Yong Xu
- Center of Chemical Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, P.R. China
| | - Fu Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| |
Collapse
|
25
|
Boyce M, Meyer B, Grulke C, Lizarraga L, Patlewicz G. Comparing the performance and coverage of selected in silico (liver) metabolism tools relative to reported studies in the literature to inform analogue selection in read-across: A case study. COMPUTATIONAL TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 21:1-15. [PMID: 35386221 PMCID: PMC8979226 DOI: 10.1016/j.comtox.2021.100208] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Changes in the regulatory landscape of chemical safety assessment call for the use of New Approach Methodologies (NAMs) including read-across to fill data gaps. One critical aspect of analogue evaluation is the extent to which target and source analogues are metabolically similar. In this study, a set of 37 structurally diverse chemicals were compiled from the EPA ToxCast inventory to compare and contrast a selection of metabolism in silico tools, in terms of their coverage and performance relative to metabolism information reported in the literature. The aim was to build understanding of the scope and capabilities of these tools and how they could be utilised in a read-across assessment. The tools were Systematic Generation of Metabolites (SyGMa), Meteor Nexus, BioTransformer, Tissue Metabolism Simulator (TIMES), OECD Toolbox, and Chemical Transformation Simulator (CTS). Performance was characterised by sensitivity and precision determined by comparing predictions against literature reported metabolites (from 44 publications). A coverage score was derived to provide a relative quantitative comparison between the tools. Meteor, TIMES, Toolbox, and CTS predictions were run in batch mode, using default settings. SyGMa and BioTransformer were run with user-defined settings, (two passes of phase I and one pass of phase II). Hierarchical clustering revealed high similarity between TIMES and Toolbox. SyGMa had the highest coverage, matching an average of 38.63% of predictions generated by the other tools though was prone to significant overprediction. It generated 5,125 metabolites, which represented 54.67% of all predictions. Precision and sensitivity values ranged from 1.1-29% and 14.7-28.3% respectively. The Toolbox had the highest performance overall. A case study was presented for 3,4-Toluenediamine (3,4-TDA), assessed for the derivation of screening-level Provisional Peer Reviewed Toxicity Values (PPRTVs), was used to demonstrate the practical role in silico metabolism information can play in analogue evaluation as part of a read-across approach.
Collapse
Affiliation(s)
- Matthew Boyce
- Oak Ridge Associated University, Oak Ridge, TN, 37830, USA
- Center for Computational Toxicology & Exposure (CCTE), U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC, 27709, USA
| | - Brian Meyer
- Center for Computational Toxicology & Exposure (CCTE), U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC, 27709, USA
| | - Chris Grulke
- Center for Computational Toxicology & Exposure (CCTE), U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC, 27709, USA
| | - Lucina Lizarraga
- Center for Public Human Health and Environmental Assessment (CPHEA), U.S. Environmental Protection Agency, Cincinnati, OH, USA
| | - Grace Patlewicz
- Center for Computational Toxicology & Exposure (CCTE), U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC, 27709, USA
| |
Collapse
|
26
|
Goldwaser E, Laurent C, Lagarde N, Fabrega S, Nay L, Villoutreix BO, Jelsch C, Nicot AB, Loriot MA, Miteva MA. Machine learning-driven identification of drugs inhibiting cytochrome P450 2C9. PLoS Comput Biol 2022; 18:e1009820. [PMID: 35081108 PMCID: PMC8820617 DOI: 10.1371/journal.pcbi.1009820] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 02/07/2022] [Accepted: 01/10/2022] [Indexed: 11/19/2022] Open
Abstract
Cytochrome P450 2C9 (CYP2C9) is a major drug-metabolizing enzyme that represents 20% of the hepatic CYPs and is responsible for the metabolism of 15% of drugs. A general concern in drug discovery is to avoid the inhibition of CYP leading to toxic drug accumulation and adverse drug-drug interactions. However, the prediction of CYP inhibition remains challenging due to its complexity. We developed an original machine learning approach for the prediction of drug-like molecules inhibiting CYP2C9. We created new predictive models by integrating CYP2C9 protein structure and dynamics knowledge, an original selection of physicochemical properties of CYP2C9 inhibitors, and machine learning modeling. We tested the machine learning models on publicly available data and demonstrated that our models successfully predicted CYP2C9 inhibitors with an accuracy, sensitivity and specificity of approximately 80%. We experimentally validated the developed approach and provided the first identification of the drugs vatalanib, piriqualone, ticagrelor and cloperidone as strong inhibitors of CYP2C9 with IC values <18 μM and sertindole, asapiprant, duvelisib and dasatinib as moderate inhibitors with IC50 values between 40 and 85 μM. Vatalanib was identified as the strongest inhibitor with an IC50 value of 0.067 μM. Metabolism assays allowed the characterization of specific metabolites of abemaciclib, cloperidone, vatalanib and tarafenacin produced by CYP2C9. The obtained results demonstrate that such a strategy could improve the prediction of drug-drug interactions in clinical practice and could be utilized to prioritize drug candidates in drug discovery pipelines.
Collapse
Affiliation(s)
- Elodie Goldwaser
- INSERM U1268 « Medicinal Chemistry and Translational Research », UMR 8038 CiTCoM, CNRS—University of Paris, Paris, France
| | | | - Nathalie Lagarde
- Laboratoire GBCM, EA7528, Conservatoire National des Arts et Métiers, 2 Rue Conté, Hésam Université, Paris, France
| | - Sylvie Fabrega
- Viral Vector for Gene Transfer core facility, Université de Paris—Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris, France
| | - Laure Nay
- Viral Vector for Gene Transfer core facility, Université de Paris—Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris, France
| | | | | | - Arnaud B. Nicot
- INSERM, Nantes Université, Center for Research in Transplantation and Translational Immunology, UMR 1064, ITUN, Nantes, France
| | - Marie-Anne Loriot
- University of Paris, INSERM U1138, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Biochimie, Paris, France
| | - Maria A. Miteva
- INSERM U1268 « Medicinal Chemistry and Translational Research », UMR 8038 CiTCoM, CNRS—University of Paris, Paris, France
| |
Collapse
|
27
|
Machine learning & deep learning in data-driven decision making of drug discovery and challenges in high-quality data acquisition in the pharmaceutical industry. Future Med Chem 2021; 14:245-270. [PMID: 34939433 DOI: 10.4155/fmc-2021-0243] [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: 01/10/2023] Open
Abstract
Predicting novel small molecule bioactivities for the target deconvolution, hit-to-lead optimization in drug discovery research, requires molecular representation. Previous reports have demonstrated that machine learning (ML) and deep learning (DL) have substantial implications in virtual screening, peptide synthesis, drug ADMET screening and biomarker discovery. These strategies can increase the positive outcomes in the drug discovery process without false-positive rates and can be achieved in a cost-effective way with a minimum duration of time by high-quality data acquisition. This review substantially discusses the recent updates in AI tools as cheminformatics application in medicinal chemistry for the data-driven decision making of drug discovery and challenges in high-quality data acquisition in the pharmaceutical industry while improving small-molecule bioactivities and properties.
Collapse
|
28
|
Phenolic-protein interactions in foods and post ingestion: Switches empowering health outcomes. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.08.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
29
|
Bassan A, Alves VM, Amberg A, Anger LT, Auerbach S, Beilke L, Bender A, Cronin MT, Cross KP, Hsieh JH, Greene N, Kemper R, Kim MT, Mumtaz M, Noeske T, Pavan M, Pletz J, Russo DP, Sabnis Y, Schaefer M, Szabo DT, Valentin JP, Wichard J, Williams D, Woolley D, Zwickl C, Myatt GJ. In silico approaches in organ toxicity hazard assessment: current status and future needs in predicting liver toxicity. COMPUTATIONAL TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 20:100187. [PMID: 35340402 PMCID: PMC8955833 DOI: 10.1016/j.comtox.2021.100187] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
Hepatotoxicity is one of the most frequently observed adverse effects resulting from exposure to a xenobiotic. For example, in pharmaceutical research and development it is one of the major reasons for drug withdrawals, clinical failures, and discontinuation of drug candidates. The development of faster and cheaper methods to assess hepatotoxicity that are both more sustainable and more informative is critically needed. The biological mechanisms and processes underpinning hepatotoxicity are summarized and experimental approaches to support the prediction of hepatotoxicity are described, including toxicokinetic considerations. The paper describes the increasingly important role of in silico approaches and highlights challenges to the adoption of these methods including the lack of a commonly agreed upon protocol for performing such an assessment and the need for in silico solutions that take dose into consideration. A proposed framework for the integration of in silico and experimental information is provided along with a case study describing how computational methods have been used to successfully respond to a regulatory question concerning non-genotoxic impurities in chemically synthesized pharmaceuticals.
Collapse
Affiliation(s)
- Arianna Bassan
- Innovatune srl, Via Giulio Zanon 130/D, 35129 Padova, Italy
| | - Vinicius M. Alves
- The National Institute of Environmental Health Sciences, Division of the National Toxicology, Program, Research Triangle Park, NC 27709, USA
| | - Alexander Amberg
- Sanofi, R&D Preclinical Safety Frankfurt, Industriepark Hoechst, D-65926 Frankfurt am Main, Germany
| | | | - Scott Auerbach
- The National Institute of Environmental Health Sciences, Division of the National Toxicology, Program, Research Triangle Park, NC 27709, USA
| | - Lisa Beilke
- Toxicology Solutions Inc., San Diego, CA, USA
| | - Andreas Bender
- AI and Data Analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW
| | - Mark T.D. Cronin
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, L3 3AF, UK
| | | | - Jui-Hua Hsieh
- The National Institute of Environmental Health Sciences, Division of the National Toxicology, Program, Research Triangle Park, NC 27709, USA
| | - Nigel Greene
- Data Science and AI, DSM, IMED Biotech Unit, AstraZeneca, Boston, USA
| | - Raymond Kemper
- Nuvalent, One Broadway, 14th floor, Cambridge, MA, 02142, USA
| | - Marlene T. Kim
- US Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, MD, 20993, USA
| | - Moiz Mumtaz
- Office of the Associate Director for Science (OADS), Agency for Toxic Substances and Disease, Registry, US Department of Health and Human Services, Atlanta, GA, USA
| | - Tobias Noeske
- Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Manuela Pavan
- Innovatune srl, Via Giulio Zanon 130/D, 35129 Padova, Italy
| | - Julia Pletz
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, L3 3AF, UK
| | - Daniel P. Russo
- Department of Chemistry, Rutgers University, Camden, NJ 08102, USA
- The Rutgers Center for Computational and Integrative Biology, Camden, NJ 08102, USA
| | - Yogesh Sabnis
- UCB Biopharma SRL, Chemin du Foriest – B-1420 Braine-l’Alleud, Belgium
| | - Markus Schaefer
- Sanofi, R&D Preclinical Safety Frankfurt, Industriepark Hoechst, D-65926 Frankfurt am Main, Germany
| | | | | | - Joerg Wichard
- Bayer AG, Genetic Toxicology, Müllerstr. 178, 13353 Berlin, Germany
| | - Dominic Williams
- Functional & Mechanistic Safety, Clinical Pharmacology & Safety Sciences, AstraZeneca, Darwin Building 310, Cambridge Science Park, Milton Rd, Cambridge CB4 0FZ, UK
| | - David Woolley
- ForthTox Limited, PO Box 13550, Linlithgow, EH49 7YU, UK
| | - Craig Zwickl
- Transendix LLC, 1407 Moores Manor, Indianapolis, IN 46229, USA
| | | |
Collapse
|
30
|
Bender BJ, Gahbauer S, Luttens A, Lyu J, Webb CM, Stein RM, Fink EA, Balius TE, Carlsson J, Irwin JJ, Shoichet BK. A practical guide to large-scale docking. Nat Protoc 2021; 16:4799-4832. [PMID: 34561691 PMCID: PMC8522653 DOI: 10.1038/s41596-021-00597-z] [Citation(s) in RCA: 269] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 06/22/2021] [Indexed: 02/08/2023]
Abstract
Structure-based docking screens of large compound libraries have become common in early drug and probe discovery. As computer efficiency has improved and compound libraries have grown, the ability to screen hundreds of millions, and even billions, of compounds has become feasible for modest-sized computer clusters. This allows the rapid and cost-effective exploration and categorization of vast chemical space into a subset enriched with potential hits for a given target. To accomplish this goal at speed, approximations are used that result in undersampling of possible configurations and inaccurate predictions of absolute binding energies. Accordingly, it is important to establish controls, as are common in other fields, to enhance the likelihood of success in spite of these challenges. Here we outline best practices and control docking calculations that help evaluate docking parameters for a given target prior to undertaking a large-scale prospective screen, with exemplification in one particular target, the melatonin receptor, where following this procedure led to direct docking hits with activities in the subnanomolar range. Additional controls are suggested to ensure specific activity for experimentally validated hit compounds. These guidelines should be useful regardless of the docking software used. Docking software described in the outlined protocol (DOCK3.7) is made freely available for academic research to explore new hits for a range of targets.
Collapse
Affiliation(s)
- Brian J Bender
- Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA, USA
| | - Stefan Gahbauer
- Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA, USA
| | - Andreas Luttens
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Jiankun Lyu
- Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA, USA
| | - Chase M Webb
- Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA, USA
| | - Reed M Stein
- Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA, USA
| | - Elissa A Fink
- Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA, USA
| | - Trent E Balius
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc, Frederick, MD, USA
| | - Jens Carlsson
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - John J Irwin
- Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA, USA
| | - Brian K Shoichet
- Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA, USA.
| |
Collapse
|
31
|
MetaClass, a Comprehensive Classification System for Predicting the Occurrence of Metabolic Reactions Based on the MetaQSAR Database. Molecules 2021; 26:molecules26195857. [PMID: 34641400 PMCID: PMC8512547 DOI: 10.3390/molecules26195857] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 11/25/2022] Open
Abstract
(1) Background: Machine learning algorithms are finding fruitful applications in predicting the ADME profile of new molecules, with a particular focus on metabolism predictions. However, the development of comprehensive metabolism predictors is hampered by the lack of highly accurate metabolic resources. Hence, we recently proposed a manually curated metabolic database (MetaQSAR), the level of accuracy of which is well suited to the development of predictive models. (2) Methods: MetaQSAR was used to extract datasets to predict the metabolic reactions subdivided into major classes, classes and subclasses. The collected datasets comprised a total of 3788 first-generation metabolic reactions. Predictive models were developed by using standard random forest algorithms and sets of physicochemical, stereo-electronic and constitutional descriptors. (3) Results: The developed models showed satisfactory performance, especially for hydrolyses and conjugations, while redox reactions were predicted with greater difficulty, which was reasonable as they depend on many complex features that are not properly encoded by the included descriptors. (4) Conclusions: The generated models allowed a precise comparison of the propensity of each metabolic reaction to be predicted and the factors affecting their predictability were discussed in detail. Overall, the study led to the development of a freely downloadable global predictor, MetaClass, which correctly predicts 80% of the reported reactions, as assessed by an explorative validation analysis on an external dataset, with an overall MCC = 0.44.
Collapse
|
32
|
Plonka W, Stork C, Šícho M, Kirchmair J. CYPlebrity: Machine learning models for the prediction of inhibitors of cytochrome P450 enzymes. Bioorg Med Chem 2021; 46:116388. [PMID: 34488021 DOI: 10.1016/j.bmc.2021.116388] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/19/2021] [Accepted: 08/24/2021] [Indexed: 10/20/2022]
Abstract
The vast majority of approved drugs are metabolized by the five major cytochrome P450 (CYP) isozymes, 1A2, 2C9, 2C19, 2D6 and 3A4. Inhibition of CYP isozymes can cause drug-drug interactions with severe pharmacological and toxicological consequences. Computational methods for the fast and reliable prediction of the inhibition of CYP isozymes by small molecules are therefore of high interest and relevance to pharmaceutical companies and a host of other industries, including the cosmetics and agrochemical industries. Today, a large number of machine learning models for predicting the inhibition of the major CYP isozymes by small molecules are available. With this work we aim to go beyond the coverage of existing models, by combining data from several major public and proprietary sources. More specifically, we used up to 18815 compounds with measured bioactivities to train random forest classification models for the individual CYP isozymes. A major advantage of the new data collection over existing ones is the better representation of the minority class, the CYP inhibitors. With the new data collection we achieved inhibitor-to-non-inhibitor ratios in the order of 1:1 (CYP1A2) to 1:3 (CYP2D6). We show that our models reach competitive performance on external data, with Matthews correlation coefficients (MCCs) ranging from 0.62 (CYP2C19) to 0.70 (CYP2D6), and areas under the receiver operating characteristic curve (AUCs) between 0.89 (CYP2C19) and 0.92 (CYPs 2D6 and 3A4). Importantly, the models show a high level of robustness, reflected in a good predictivity also for compounds that are structurally dissimilar to the compounds represented in the training data. The best models presented in this work are freely accessible for academic research via a web service.
Collapse
Affiliation(s)
- Wojciech Plonka
- Universität Hamburg, Center for Bioinformatics (ZBH), Hamburg, Bundesstr. 43, 20146, Germany; FQS Poland (Fujitsu Group), Parkowa 11, 30-538 Cracow, Poland
| | - Conrad Stork
- Universität Hamburg, Center for Bioinformatics (ZBH), Hamburg, Bundesstr. 43, 20146, Germany
| | - Martin Šícho
- CZ-OPENSCREEN: National Infrastructure for Chemical Biology, Department of Informatics and Chemistry, Faculty of Chemical Technology, University of Chemistry and Technology Prague, Technická 5, 166 28, Prague, Czech Republic
| | - Johannes Kirchmair
- Universität Hamburg, Center for Bioinformatics (ZBH), Hamburg, Bundesstr. 43, 20146, Germany; Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Althanstr. 14, 1090 Vienna, Austria.
| |
Collapse
|
33
|
Holmer M, de Bruyn Kops C, Stork C, Kirchmair J. CYPstrate: A Set of Machine Learning Models for the Accurate Classification of Cytochrome P450 Enzyme Substrates and Non-Substrates. Molecules 2021; 26:molecules26154678. [PMID: 34361831 PMCID: PMC8347321 DOI: 10.3390/molecules26154678] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 11/16/2022] Open
Abstract
The interaction of small organic molecules such as drugs, agrochemicals, and cosmetics with cytochrome P450 enzymes (CYPs) can lead to substantial changes in the bioavailability of active substances and hence consequences with respect to pharmacological efficacy and toxicity. Therefore, efficient means of predicting the interactions of small organic molecules with CYPs are of high importance to a host of different industries. In this work, we present a new set of machine learning models for the classification of xenobiotics into substrates and non-substrates of nine human CYP isozymes: CYPs 1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, and 3A4. The models are trained on an extended, high-quality collection of known substrates and non-substrates and have been subjected to thorough validation. Our results show that the models yield competitive performance and are favorable for the detection of CYP substrates. In particular, a new consensus model reached high performance, with Matthews correlation coefficients (MCCs) between 0.45 (CYP2C8) and 0.85 (CYP3A4), although at the cost of coverage. The best models presented in this work are accessible free of charge via the "CYPstrate" module of the New E-Resource for Drug Discovery (NERDD).
Collapse
Affiliation(s)
- Malte Holmer
- Center for Bioinformatics (ZBH), Department of Informatics, Universität Hamburg, 20146 Hamburg, Germany; (M.H.); (C.d.B.K.); (C.S.)
| | - Christina de Bruyn Kops
- Center for Bioinformatics (ZBH), Department of Informatics, Universität Hamburg, 20146 Hamburg, Germany; (M.H.); (C.d.B.K.); (C.S.)
| | - Conrad Stork
- Center for Bioinformatics (ZBH), Department of Informatics, Universität Hamburg, 20146 Hamburg, Germany; (M.H.); (C.d.B.K.); (C.S.)
| | - Johannes Kirchmair
- Center for Bioinformatics (ZBH), Department of Informatics, Universität Hamburg, 20146 Hamburg, Germany; (M.H.); (C.d.B.K.); (C.S.)
- Division of Pharmaceutical Chemistry, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria
- Correspondence:
| |
Collapse
|
34
|
Zhang X, Xu M, Wu Z, Liu G, Tang Y, Li W. Assessment of CYP2C9 Structural Models for Site of Metabolism Prediction. ChemMedChem 2021; 16:1754-1763. [PMID: 33600055 DOI: 10.1002/cmdc.202000964] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/07/2021] [Indexed: 11/07/2022]
Abstract
Structure-based prediction of a compound's potential sites of metabolism (SOMs) mediated by cytochromes P450 (CYPs) is highly advantageous in the early stage of drug discovery. However, the accuracy of the SOMs prediction can be influenced by several factors. CYP2C9 is one of the major drug-metabolizing enzymes in humans and is responsible for the metabolism of ∼13 % of clinically used drugs. In this study, we systematically evaluated the effects of protein crystal structure models, scoring functions, heme forms, conserved active-site water molecules, and protein flexibility on SOMs prediction of CYP2C9 substrates. Our results demonstrated that, on average, ChemScore and GlideScore outperformed four other scoring functions: Vina, GoldScore, ChemPLP, and ASP. The performance of the crystal structure models with pentacoordinated heme was generally superior to that of the hexacoordinated iron-oxo heme (referred to as Compound I) models. Inclusion of the conserved active-site water molecule improved the prediction accuracy of GlideScore, but reduced the accuracy of ChemScore. In addition, the effect of the conserved water on SOMs prediction was found to be dependent on the receptor model and the substrate. We further found that one of snapshots from molecular dynamics simulations on the apo form can improve the prediction accuracy when compared to the crystal structural model.
Collapse
Affiliation(s)
- Xiaoxiao Zhang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 20023, P. R. China
| | - Minjie Xu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 20023, P. R. China
| | - Zengrui Wu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 20023, P. R. China
| | - Guixia Liu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 20023, P. R. China
| | - Yun Tang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 20023, P. R. China
| | - Weihua Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 20023, P. R. China
| |
Collapse
|
35
|
Molecular probes for human cytochrome P450 enzymes: Recent progress and future perspectives. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2020.213600] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
36
|
Park RM. A Simple Toxicokinetic Model Exhibiting Complex Dynamics and Nonlinear Exposure Response. RISK ANALYSIS : AN OFFICIAL PUBLICATION OF THE SOCIETY FOR RISK ANALYSIS 2020; 40:2561-2571. [PMID: 32632964 PMCID: PMC7748990 DOI: 10.1111/risa.13547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 06/02/2020] [Accepted: 06/13/2020] [Indexed: 06/11/2023]
Abstract
Uncertainty in model predictions of exposure response at low exposures is a problem for risk assessment. A particular interest is the internal concentration of an agent in biological systems as a function of external exposure concentrations. Physiologically based pharmacokinetic (PBPK) models permit estimation of internal exposure concentrations in target tissues but most assume that model parameters are either fixed or instantaneously dose-dependent. Taking into account response times for biological regulatory mechanisms introduces new dynamic behaviors that have implications for low-dose exposure response in chronic exposure. A simple one-compartment simulation model is described in which internal concentrations summed over time exhibit significant nonlinearity and nonmonotonicity in relation to external concentrations due to delayed up- or downregulation of a metabolic pathway. These behaviors could be the mechanistic basis for homeostasis and for some apparent hormetic effects.
Collapse
Affiliation(s)
- Robert M. Park
- Division of Science Integration, National Institute for Occupational Safety and Health, 1090 Tusculum Ave, MS C-15, Cincinnati OH, USA
| |
Collapse
|
37
|
Wu F, Zhou Y, Li L, Shen X, Chen G, Wang X, Liang X, Tan M, Huang Z. Computational Approaches in Preclinical Studies on Drug Discovery and Development. Front Chem 2020; 8:726. [PMID: 33062633 PMCID: PMC7517894 DOI: 10.3389/fchem.2020.00726] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 07/14/2020] [Indexed: 12/11/2022] Open
Abstract
Because undesirable pharmacokinetics and toxicity are significant reasons for the failure of drug development in the costly late stage, it has been widely recognized that drug ADMET properties should be considered as early as possible to reduce failure rates in the clinical phase of drug discovery. Concurrently, drug recalls have become increasingly common in recent years, prompting pharmaceutical companies to increase attention toward the safety evaluation of preclinical drugs. In vitro and in vivo drug evaluation techniques are currently more mature in preclinical applications, but these technologies are costly. In recent years, with the rapid development of computer science, in silico technology has been widely used to evaluate the relevant properties of drugs in the preclinical stage and has produced many software programs and in silico models, further promoting the study of ADMET in vitro. In this review, we first introduce the two ADMET prediction categories (molecular modeling and data modeling). Then, we perform a systematic classification and description of the databases and software commonly used for ADMET prediction. We focus on some widely studied ADMT properties as well as PBPK simulation, and we list some applications that are related to the prediction categories and web tools. Finally, we discuss challenges and limitations in the preclinical area and propose some suggestions and prospects for the future.
Collapse
Affiliation(s)
- Fengxu Wu
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Research Platform Service Management Center, Dongguan, China
- Key Laboratory of Pesticide & Chemical Biology, Ministry of Education, College of Chemistry, Central China Normal University, Wuhan, China
| | - Yuquan Zhou
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Research Platform Service Management Center, Dongguan, China
- The Second School of Clinical Medicine, Guangdong Medical University, Dongguan, China
| | - Langhui Li
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Research Platform Service Management Center, Dongguan, China
- Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Xianhuan Shen
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Research Platform Service Management Center, Dongguan, China
- Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Ganying Chen
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Research Platform Service Management Center, Dongguan, China
- The Second School of Clinical Medicine, Guangdong Medical University, Dongguan, China
| | - Xiaoqing Wang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Research Platform Service Management Center, Dongguan, China
- Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Xianyang Liang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Research Platform Service Management Center, Dongguan, China
- The Second School of Clinical Medicine, Guangdong Medical University, Dongguan, China
| | - Mengyuan Tan
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Research Platform Service Management Center, Dongguan, China
- Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Zunnan Huang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Research Platform Service Management Center, Dongguan, China
- Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| |
Collapse
|
38
|
Liu J, Machalz D, Wolber G, Sorensen EJ, Bureik M. New Proluciferin Substrates for Human CYP4 Family Enzymes. Appl Biochem Biotechnol 2020; 193:218-237. [PMID: 32869209 DOI: 10.1007/s12010-020-03388-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/16/2020] [Indexed: 02/08/2023]
Abstract
We report the synthesis of seven new proluciferins for convenient activity determination of enzymes belonging to the cytochrome P450 (CYP) 4 family. Biotransformation of these probe substrates was monitored using each of the twelve human CYP4 family members, and eight were found to act at least on one of them. For all substrates, activity of CYP4Z1 was always highest, while that of CYP4F8 was always second highest. Site of metabolism (SOM) predictions involving SMARTCyp and docking experiments helped to rationalize the observed activity trends linked to substrate accessibility and reactivity. We further report the first homology model of CYP4F8 including suggested substrate recognition residues in a catalytically competent conformation accessed by replica exchange solute tempering (REST) simulations.
Collapse
Affiliation(s)
- Jingyao Liu
- School of Pharmaceutical Science and Technology, Health Sciences Platform, Tianjin University, Tianjin, 300072, China
| | - David Machalz
- Pharmaceutical and Medicinal Chemistry (Computer-Aided Drug Design), Institute of Pharmacy, Freie Universität Berlin, 14195, Berlin, Germany
| | - Gerhard Wolber
- Pharmaceutical and Medicinal Chemistry (Computer-Aided Drug Design), Institute of Pharmacy, Freie Universität Berlin, 14195, Berlin, Germany
| | - Erik J Sorensen
- School of Pharmaceutical Science and Technology, Health Sciences Platform, Tianjin University, Tianjin, 300072, China. .,Department of Chemistry, Princeton University, Princeton, NJ, 08544, USA.
| | - Matthias Bureik
- School of Pharmaceutical Science and Technology, Health Sciences Platform, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
39
|
de Bruyn Kops C, Šícho M, Mazzolari A, Kirchmair J. GLORYx: Prediction of the Metabolites Resulting from Phase 1 and Phase 2 Biotransformations of Xenobiotics. Chem Res Toxicol 2020; 34:286-299. [PMID: 32786543 PMCID: PMC7887798 DOI: 10.1021/acs.chemrestox.0c00224] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Predicting
the structures of metabolites formed in humans can provide
advantageous insights for the development of drugs and other compounds.
Here we present GLORYx, which integrates machine learning-based site
of metabolism (SoM) prediction with reaction rule sets to predict
and rank the structures of metabolites that could potentially be formed
by phase 1 and/or phase 2 metabolism. GLORYx extends the approach
from our previously developed tool GLORY, which predicted metabolite
structures for cytochrome P450-mediated metabolism only. A robust
approach to ranking the predicted metabolites is attained by using
the SoM probabilities predicted by the FAME 3 machine learning models
to score the predicted metabolites. On a manually curated test data
set containing both phase 1 and phase 2 metabolites, GLORYx achieves
a recall of 77% and an area under the receiver operating characteristic
curve (AUC) of 0.79. Separate analysis of performance on a large amount
of freely available phase 1 and phase 2 metabolite data indicates
that achieving a meaningful ranking of predicted metabolites is more
difficult for phase 2 than for phase 1 metabolites. GLORYx is freely
available as a web server at https://nerdd.zbh.uni-hamburg.de/ and is also provided as a software package upon request. The data
sets as well as all the reaction rules from this work are also made
freely available.
Collapse
Affiliation(s)
- Christina de Bruyn Kops
- Center for Bioinformatics (ZBH), Department of Informatics, Faculty of Mathematics, Informatics and Natural Sciences, Universität Hamburg, 20146 Hamburg, Germany
| | - Martin Šícho
- CZ-OPENSCREEN: National Infrastructure for Chemical Biology, Department of Informatics and Chemistry, Faculty of Chemical Technology, University of Chemistry and Technology Prague, 166 28 Prague 6, Czech Republic
| | - Angelica Mazzolari
- Facoltà di Scienze del Farmaco, Dipartimento di Scienze Farmaceutiche "Pietro Pratesi", Università degli Studi di Milano, I-20133 Milan, Italy
| | - Johannes Kirchmair
- Center for Bioinformatics (ZBH), Department of Informatics, Faculty of Mathematics, Informatics and Natural Sciences, Universität Hamburg, 20146 Hamburg, Germany.,Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
40
|
Assis DB, Aragão Neto HDC, da Fonsêca DV, de Andrade HHN, Braga RM, Badr N, Maia MDS, Castro RD, Scotti L, Scotti MT, de Almeida RN. Antinociceptive Activity of Chemical Components of Essential Oils That Involves Docking Studies: A Review. Front Pharmacol 2020; 11:777. [PMID: 32547391 PMCID: PMC7272657 DOI: 10.3389/fphar.2020.00777] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/11/2020] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Pain is considered an unpleasant sensory and emotional experience, being considered as one of the most important causes of human suffering. Computational chemistry associated with bioinformatics has stood out in the process of developing new drugs, through natural products, to manage this condition. OBJECTIVE To analyze, through literature data, recent molecular coupling studies on the antinociceptive activity of essential oils and monoterpenes. DATA SOURCE Systematic search of the literature considering the years of publications between 2005 and December 2019, in the electronic databases PubMed and Science Direct. ELIGIBILITY CRITERIA Were considered as criteria of 1) Biological activity: non-clinical effects of an OE and/or monoterpenes on antinociceptive activity based on animal models and in silico analysis, 2) studies with plant material: chemically characterized essential oils and/or their constituents isolated, 3) clinical and non-clinical studies with in silico analysis to assess antinociceptive activity, 4) articles published in English. Exclusion criteria were literature review, report or case series, meta-analysis, theses, dissertations, and book chapter. RESULTS Of 16,006 articles, 16 articles fulfilled all the criteria. All selected studies were non-clinical. The most prominent plant families used were Asteraceae, Euphorbiaceae, Verbenaceae, Lamiaceae, and Lauraceae. Among the phytochemicals studied were α-Terpineol, 3-(5-substituted-1,3,4-oxadiazol-2-yl)-N'-[2-oxo-1,2-dihydro-3H-indol-3-ylidene] propane hydrazide, β-cyclodextrin complexed with citronellal, (-)-α-bisabolol, β-cyclodextrin complexed with farnesol, and p-Cymene. The softwares used for docking studies were Molegro Virtual Docker, Sybyl®X, Vlife MDS, AutoDock Vina, Hex Protein Docking, and AutoDock 4.2 in PyRx 0.9. The molecular targets/complexes used were Nitric Oxide Synthase, COX-2, GluR2-S1S2, TRPV1, β-CD complex, CaV1, CaV2.1, CaV2.2, and CaV2.3, 5-HT receptor, delta receptor, kappa receptor, and MU (μ) receptor, alpha adrenergic, opioid, and serotonergic receptors, muscarinic receptors and GABAA opioid and serotonin receptors, 5-HT3 and M2 receptors. Many of the covered studies used molecular coupling to investigate the mechanism of action of various compounds, as well as molecular dynamics to investigate the stability of protein-ligand complexes. CONCLUSIONS The studies revealed that through the advancement of more robust computational techniques that complement the experimental studies, they may allow some notes on the identification of a new candidate molecule for therapeutic use.
Collapse
Affiliation(s)
- Davidson Barbosa Assis
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, João Pessoa, Brazil
| | | | - Diogo Vilar da Fonsêca
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, João Pessoa, Brazil
| | - Humberto Hugo Nunes de Andrade
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, João Pessoa, Brazil
| | - Renan Marinho Braga
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, João Pessoa, Brazil
| | - Nader Badr
- First Faculty of Medicine, Charles University, Prague, Czechia
| | - Mayara dos Santos Maia
- Cheminformatics Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, João Pessoa, Brazil
| | - Ricardo Dias Castro
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, João Pessoa, Brazil
| | - Luciana Scotti
- Cheminformatics Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, João Pessoa, Brazil
| | - Marcus Tullius Scotti
- Cheminformatics Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, João Pessoa, Brazil
| | - Reinaldo Nóbrega de Almeida
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, João Pessoa, Brazil
| |
Collapse
|
41
|
Russell LE, Schleiff MA, Gonzalez E, Bart AG, Broccatelli F, Hartman JH, Humphreys WG, Lauschke VM, Martin I, Nwabufo C, Prasad B, Scott EE, Segall M, Takahashi R, Taub ME, Sodhi JK. Advances in the study of drug metabolism - symposium report of the 12th Meeting of the International Society for the Study of Xenobiotics (ISSX). Drug Metab Rev 2020; 52:395-407. [PMID: 32456484 DOI: 10.1080/03602532.2020.1765793] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The 12th International Society for the Study of Xenobiotics (ISSX) meeting, held in Portland, OR, USA from July 28 to 31, 2019, was attended by diverse members of the pharmaceutical sciences community. The ISSX New Investigators Group provides learning and professional growth opportunities for student and early career members of ISSX. To share meeting content with those who were unable to attend, the ISSX New Investigators herein elected to highlight the "Advances in the Study of Drug Metabolism" symposium, as it engaged attendees with diverse backgrounds. This session covered a wide range of current topics in drug metabolism research including predicting sites and routes of metabolism, metabolite identification, ligand docking, and medicinal and natural products chemistry, and highlighted approaches complemented by computational modeling. In silico tools have been increasingly applied in both academic and industrial settings, alongside traditional and evolving in vitro techniques, to strengthen and streamline pharmaceutical research. Approaches such as quantum mechanics simulations facilitate understanding of reaction energetics toward prediction of routes and sites of drug metabolism. Furthermore, in tandem with crystallographic and orthogonal wet lab techniques for structural validation of drug metabolizing enzymes, in silico models can aid understanding of substrate recognition by particular enzymes, identify metabolic soft spots and predict toxic metabolites for improved molecular design. Of note, integration of chemical synthesis and biosynthesis using natural products remains an important approach for identifying new chemical scaffolds in drug discovery. These subjects, compiled by the symposium organizers, presenters, and the ISSX New Investigators Group, are discussed in this review.
Collapse
Affiliation(s)
- Laura E Russell
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | - Mary Alexandra Schleiff
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Eric Gonzalez
- Division of Pre-Clinical Innovation, Therapeutic Development Branch, National Center for Advancing Translational Sciences, Bethesda, MD, USA.,Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Aaron G Bart
- Program in Biophysics, University of Michigan, Ann Arbor, MI, USA
| | - Fabio Broccatelli
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Jessica H Hartman
- Nicholas School of the Environment, Duke University, Durham, NC, USA
| | | | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Bhagwat Prasad
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, USA
| | - Emily E Scott
- Program in Biophysics, University of Michigan, Ann Arbor, MI, USA.,Department of Medicinal Chemistry and Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Mitchell E Taub
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, USA
| | - Jasleen K Sodhi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
42
|
Luirink RA, Verkade‐Vreeker MCA, Commandeur JNM, Geerke DP. A Modified Arrhenius Approach to Thermodynamically Study Regioselectivity in Cytochrome P450-Catalyzed Substrate Conversion. Chembiochem 2020; 21:1461-1472. [PMID: 31919943 PMCID: PMC7318578 DOI: 10.1002/cbic.201900751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Indexed: 12/21/2022]
Abstract
The regio- (and stereo-)selectivity and specific activity of cytochrome P450s are determined by the accessibility of potential sites of metabolism (SOMs) of the bound substrate relative to the heme, and the activation barrier of the regioselective oxidation reaction(s). The accessibility of potential SOMs depends on the relative binding free energy (ΔΔGbind ) of the catalytically active substrate-binding poses, and the probability of the substrate to adopt a transition-state geometry. An established experimental method to measure activation energies of enzymatic reactions is the analysis of reaction rate constants at different temperatures and the construction of Arrhenius plots. This is a challenge for multistep P450-catalyzed processes that involve redox partners. We introduce a modified Arrhenius approach to overcome the limitations in studying P450 selectivity, which can be applied in multiproduct enzyme catalysis. Our approach gives combined information on relative activation energies, ΔΔGbind values, and collision entropies, yielding direct insight into the basis of selectivity in substrate conversion.
Collapse
Affiliation(s)
- Rosa A. Luirink
- AIMMS Division of Molecular ToxicologyVrije UniversiteitDe Boelelaan 11081081 HZAmsterdamThe Netherlands
| | | | - Jan N. M. Commandeur
- AIMMS Division of Molecular ToxicologyVrije UniversiteitDe Boelelaan 11081081 HZAmsterdamThe Netherlands
| | - Daan P. Geerke
- AIMMS Division of Molecular ToxicologyVrije UniversiteitDe Boelelaan 11081081 HZAmsterdamThe Netherlands
| |
Collapse
|
43
|
Holmes JB, Doh CY, Mamidi R, Li J, Stelzer JE. Strategies for targeting the cardiac sarcomere: avenues for novel drug discovery. Expert Opin Drug Discov 2020; 15:457-469. [PMID: 32067508 PMCID: PMC7065952 DOI: 10.1080/17460441.2020.1722637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/24/2020] [Indexed: 01/10/2023]
Abstract
Introduction: Heart failure remains one of the largest clinical challenges in the United States. Researchers have continually searched for more effective heart failure treatments that target the cardiac sarcomere but have found few successes despite numerous expensive cardiovascular clinical trials. Among many reasons, the high failure rate of cardiovascular clinical trials may be partly due to incomplete characterization of a drug candidate's complex interaction with cardiac physiology.Areas covered: In this review, the authors address the issue of preclinical cardiovascular studies of sarcomere-targeting heart failure therapies. The authors consider inherent tradeoffs made between mechanistic transparency and physiological fidelity for several relevant preclinical techniques at the atomic, molecular, heart muscle fiber, whole heart, and whole-organism levels. Thus, the authors suggest a comprehensive, bottom-up approach to preclinical cardiovascular studies that fosters scientific rigor and hypothesis-driven drug discovery.Expert opinion: In the authors' opinion, the implementation of hypothesis-driven drug discovery practices, such as the bottom-up approach to preclinical cardiovascular studies, will be imperative for the successful development of novel heart failure treatments. However, additional changes to clinical definitions of heart failure and current drug discovery culture must accompany the bottom-up approach to maximize the effectiveness of hypothesis-driven drug discovery.
Collapse
Affiliation(s)
- Joshua B Holmes
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Chang Yoon Doh
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Ranganath Mamidi
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Jiayang Li
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Julian E Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
44
|
Fine J, Konc J, Samudrala R, Chopra G. CANDOCK: Chemical Atomic Network-Based Hierarchical Flexible Docking Algorithm Using Generalized Statistical Potentials. J Chem Inf Model 2020; 60:1509-1527. [PMID: 32069042 PMCID: PMC12034428 DOI: 10.1021/acs.jcim.9b00686] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Small-molecule docking has proven to be invaluable for drug design and discovery. However, existing docking methods have several limitations such as improper treatment of the interactions of essential components in the chemical environment of the binding pocket (e.g., cofactors, metal ions, etc.), incomplete sampling of chemically relevant ligand conformational space, and the inability to consistently correlate docking scores of the best binding pose with experimental binding affinities. We present CANDOCK, a novel docking algorithm, that utilizes a hierarchical approach to reconstruct ligands from an atomic grid using graph theory and generalized statistical potential functions to sample biologically relevant ligand conformations. Our algorithm accounts for protein flexibility, solvent, metal ions, and cofactor interactions in the binding pocket that are traditionally ignored by current methods. We evaluate the algorithm on the PDBbind, Astex, and PINC proteins to show its ability to reproduce the binding mode of the ligands that is independent of the initial ligand conformation in these benchmarks. Finally, we identify the best selector and ranker potential functions such that the statistical score of the best selected docked pose correlates with the experimental binding affinities of the ligands for any given protein target. Our results indicate that CANDOCK is a generalized flexible docking method that addresses several limitations of current docking methods by considering all interactions in the chemical environment of a binding pocket for correlating the best-docked pose with biological activity. CANDOCK along with all structures and scripts used for benchmarking is available at https://github.com/chopralab/candock_benchmark.
Collapse
Affiliation(s)
- Jonathan Fine
- Department of Chemistry, Purdue University, 720 Clinic Drive, West Lafayette, IN, USA 47906
| | - Janez Konc
- National Institute of Chemistry, Hajdrihova 19, SI−1000, Ljubljana, Slovenia
| | - Ram Samudrala
- Department of Biomedical Informatics, SUNY, Buffalo, NY, USA 14260
| | - Gaurav Chopra
- Department of Chemistry, Purdue University, 720 Clinic Drive, West Lafayette, IN, USA 47906
- Purdue Institute for Drug Discovery
- Purdue Center for Cancer Research
- Purdue Institute for Inflammation, Immunology and Infectious Disease
- Purdue Institute for Integrative Neuroscience
- Integrative Data Science Initiative
| |
Collapse
|
45
|
Rifai EA, Ferrario V, Pleiss J, Geerke DP. Combined Linear Interaction Energy and Alchemical Solvation Free-Energy Approach for Protein-Binding Affinity Computation. J Chem Theory Comput 2020; 16:1300-1310. [PMID: 31894691 PMCID: PMC7017367 DOI: 10.1021/acs.jctc.9b00890] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Calculating free energies of binding (ΔGbind) between ligands and their target protein is of major interest to drug discovery and safety, yet it is still associated with several challenges and difficulties. Linear interaction energy (LIE) is an efficient in silico method for ΔGbind computation. LIE models can be trained and used to directly calculate binding affinities from interaction energies involving ligands in the bound and unbound states only, and LIE can be combined with statistical weighting to calculate ΔGbind for flexible proteins that may bind their ligands in multiple orientations. Here, we investigate if LIE predictions can be effectively improved by explicitly including the entropy of (de)solvation into our free-energy calculations. For that purpose, we combine LIE calculations for the protein-ligand-bound state with explicit free-energy perturbation to rigorously compute the unbound ligand's solvation free energy. We show that for 28 Cytochrome P450 2A6 (CYP2A6) ligands, coupling LIE with alchemical solvation free-energy calculation helps to improve obtained correlation between computed and reference (experimental) binding data.
Collapse
Affiliation(s)
- Eko Aditya Rifai
- AIMMS Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences , Vrije Universiteit Amsterdam , De Boelelaan 1108 , 1081 HZ Amsterdam , The Netherlands
| | - Valerio Ferrario
- Institute of Biochemistry and Technical Biochemistry , Universität Stuttgart , Allmandring 31 , 70569 Stuttgart , Germany
| | - Jürgen Pleiss
- Institute of Biochemistry and Technical Biochemistry , Universität Stuttgart , Allmandring 31 , 70569 Stuttgart , Germany
| | - Daan P Geerke
- AIMMS Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences , Vrije Universiteit Amsterdam , De Boelelaan 1108 , 1081 HZ Amsterdam , The Netherlands
| |
Collapse
|
46
|
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
|
47
|
Kato H. Computational prediction of cytochrome P450 inhibition and induction. Drug Metab Pharmacokinet 2019; 35:30-44. [PMID: 31902468 DOI: 10.1016/j.dmpk.2019.11.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/27/2019] [Accepted: 11/17/2019] [Indexed: 12/14/2022]
Abstract
Cytochrome P450 (CYP) enzymes play an important role in the phase I metabolism of many xenobiotics. Most drug-drug interactions (DDIs) associated with CYP are caused by either CYP inhibition or induction. The early detection of potential DDIs is highly desirable in the pharmaceutical industry because DDIs can cause serious adverse events, which can lead to poor patient health and drug development failures. Recently, many computational studies predicting CYP inhibition and induction have been reported. The current computational modeling approaches for CYP metabolism are classified as ligand- and structure-based; various techniques, such as quantitative structure-activity relationships, machine learning, docking, and molecular dynamic simulation, are involved in both the approaches. Recently, combining these two approaches have resulted in improvements in the prediction accuracy of DDIs. In this review, we present important, recent developments in the computational prediction of the inhibition of four clinically crucial CYP isoforms (CYP1A2, 2C9, 2D6, and 3A4) and three nuclear receptors (aryl hydrocarbon receptor, constitutive androstane receptor, and pregnane X receptor) involved in the induction of CYP1A2, 2B6, and 3A4, respectively.
Collapse
Affiliation(s)
- Harutoshi Kato
- DMPK Research Laboratories, Mitsubishi Tanabe Pharma Corporation, Aoba-ku, Yokohama-shi, 227-0033, Japan.
| |
Collapse
|
48
|
Lazzara PR, Moore TW. Scaffold-hopping as a strategy to address metabolic liabilities of aromatic compounds. RSC Med Chem 2019; 11:18-29. [PMID: 33479602 DOI: 10.1039/c9md00396g] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 10/09/2019] [Indexed: 12/31/2022] Open
Abstract
Understanding and minimizing oxidative metabolism of aromatic compounds is a key hurdle in lead optimization. Metabolic processes not only clear compounds from the body, but they can also transform parent compounds into reactive metabolites. One particularly useful strategy when addressing metabolically labile or oxidation-prone structures is scaffold-hopping. Replacement of an aromatic system with a more electron-deficient ring system can often increase robustness towards cytochrome P450-mediated oxidation while conserving the structural requirements of the pharmacophore. The most common example of this substitution strategy, replacement of a phenyl ring with a pyridyl substituent, is prevalent throughout the literature; however scaffold-hopping encompasses a much wider scope of heterocycle replacement. This review will showcase recent examples where different scaffold-hopping approaches were used to reduce metabolic clearance or block the formation of reactive metabolites. Additionally, we will highlight considerations that should be made to garner the most benefit from a scaffold-hopping strategy for lead optimization.
Collapse
Affiliation(s)
- Phillip R Lazzara
- Department of Pharmaceutical Sciences , College of Pharmacy , University of Illinois at Chicago , 833 S. Wood Street , Chicago , IL 60612 , USA .
| | - Terry W Moore
- Department of Pharmaceutical Sciences , College of Pharmacy , University of Illinois at Chicago , 833 S. Wood Street , Chicago , IL 60612 , USA . .,University of Illinois Cancer Center , University of Illinois at Chicago , 1801 W. Taylor Street , Chicago , IL 60612 , USA
| |
Collapse
|
49
|
Quiroga I, Scior T. Induced fit for cytochrome P450 3A4 based on molecular dynamics. ADMET AND DMPK 2019; 7:252-266. [PMID: 35359616 PMCID: PMC8963583 DOI: 10.5599/admet.729] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/19/2019] [Indexed: 11/18/2022] Open
Abstract
The present study aims at numerically describing to what extent substrate - enzyme complexes in solution may change over time as a natural process of conformational changes for a liganded enzyme in comparison to those movements which occur independently from substrate interaction, i.e. without a ligand. To this end, we selected structurally known pairs of liganded / unliganded CYP450 3A4 enzymes with different geometries hinting at induced fit events. We carried out molecular dynamics simulations (MD) comparing the trajectories in a "cross-over" protocol: (i) we added the ligand to the unliganded crystal form which should adopt geometries similar to the known geometry of the liganded crystal structure during MD, and - conversely - (ii) we removed the bound ligand form the known liganded complex to test if a geometry similar to the known unliganded (apo-) form can be adopted during MD. To compare continues changes we measured root means square deviations and frequencies. Results for case (i) hint at larger conformational changes required for accepting the substrate during its approach to final position - in contrast to case (ii) when mobility is fairly reduced by ligand binding (strain energy). In conclusion, a larger conformational sampling prior to ligand binding and the freezing-in (rigidity) of conformations for bound ligands can be interpreted as two conditions linked to induced-fit.
Collapse
Affiliation(s)
- Israel Quiroga
- Faculty of Chemical Sciences, Benemérita Universidad Autónoma de Puebla, Puebla, Pue., Mexico
| | - Thomas Scior
- Faculty of Chemical Sciences, Benemérita Universidad Autónoma de Puebla, Puebla, Pue., Mexico
| |
Collapse
|
50
|
Saito T, Kambara H, Takano Y. Quantitative assessment of reparameterized PM6 (rPM6) for hydrogen abstraction reactions. Mol Phys 2019. [DOI: 10.1080/00268976.2019.1700313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Toru Saito
- Department of Biomedical Information Sciences, Graduate School of Information Sciences, Hiroshima City University, Hiroshima Japan
| | - Hiroki Kambara
- Department of Biomedical Information Sciences, Graduate School of Information Sciences, Hiroshima City University, Hiroshima Japan
| | - Yu Takano
- Department of Biomedical Information Sciences, Graduate School of Information Sciences, Hiroshima City University, Hiroshima Japan
| |
Collapse
|