1
|
Gomha SM, Abolibda TZ, Alruwaili AH, Farag B, Boraie WE, Al-Hussain SA, Zaki MEA, Hussein AM. Efficient Green Synthesis of Hydrazide Derivatives Using L-Proline: Structural Characterization, Anticancer Activity, and Molecular Docking Studies. Catalysts 2024; 14:489. [DOI: 10.3390/catal14080489] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025] Open
Abstract
Green synthesis using L-proline as an organocatalyst is crucial due to its reusability, mild conditions, clean reactions, easy workup, high purity, short reaction times, and high yields. However, existing methods often involve harsh conditions and longer reaction times. In this study, 2-cyano-N’-(2-cyanoacetyl)acetohydrazide (3) was prepared and condensed with various benzaldehyde derivatives to yield 2-cyano-N’-(2-cyano-3-phenylacryloyl)-3-phenylacrylohydrazide derivatives (5a–e, 7a,b) using a grinding technique with moist L-proline. Additionally, three 2-cyano-N’-(2-cyano-3-heterylbut-2-enoyl)-3-heterylbut-2-enehydrazides (9, 11, 13) were synthesized by condensing compound 3 with respective (heteraryl)ketones (8, 10, 12) following the same method. The synthesized compounds were characterized using IR, NMR, and MS spectroscopy. L-proline’s reusability was confirmed for up to four cycles without significant yield loss, showcasing the protocol’s efficiency and sustainability. The new compounds were screened for anticancer activities against the HCT-116 colon carcinoma cell line using the MTT assay. Molecular docking studies revealed the binding conformations of the most potent compounds to the target protein (PDB ID 6MTU), correlating well with in vitro results. In silico ADMET analysis indicated favorable pharmacokinetic properties, highlighting these novel compounds as promising targeted anti-colon cancer agents.
Collapse
Affiliation(s)
- Sobhi M. Gomha
- Chemistry Department, Faculty of Science, Islamic University of Madinah, Madinah 42351, Saudi Arabia
| | - Tariq Z. Abolibda
- Chemistry Department, Faculty of Science, Islamic University of Madinah, Madinah 42351, Saudi Arabia
| | - Awatif H. Alruwaili
- Department of Chemistry, Faculty of Science, Northern Border University, Arar 73222, Saudi Arabia
| | - Basant Farag
- Department of Chemistry, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
| | - Waleed E. Boraie
- Department of Chemistry, College of Science, King Faisal University, Hofuf 31982, Saudi Arabia
| | - Sami A. Al-Hussain
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Magdi E. A. Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Ahmed M. Hussein
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef 62511, Egypt
- Chemistry Department, College of Science and Humanities—Al Quwaiiyah, Shaqra University, Al-Dawadmi 11911, Saudi Arabia
| |
Collapse
|
2
|
Al-Humaidi JY, Albedair LA, Farag B, Zaki ME, Mukhrish YE, Gomha SM. Design and synthesis of novel hybrids incorporating thiadiazole or thiazole-naphthalene: Anticancer assessment and molecular docking study. RESULTS IN CHEMISTRY 2024; 7:101475. [DOI: 10.1016/j.rechem.2024.101475] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025] Open
|
3
|
Lin J, Wen L, Zhou Y, Wang S, Ye H, Su J, Li J, Shu J, Huang J, Zhou P. PepQSAR: a comprehensive data source and information platform for peptide quantitative structure-activity relationships. Amino Acids 2023; 55:235-242. [PMID: 36474016 DOI: 10.1007/s00726-022-03219-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Abstract
Peptide quantitative structure-activity relationships (pQSARs) have been widely applied to the statistical modeling and empirical prediction of peptide activity, property and feature. In the procedure, the peptide structure is characterized at sequence level using amino acid descriptors (AADs) and then correlated with observations by machine learning methods (MLMs), consequently resulting in a variety of quantitative regression models used to explain the structural factors that govern peptide activities, to generalize peptide properties of unknown from known samples, and to design new peptides with desired features. In this study, we developed a comprehensive platform, termed PepQSAR database, which is a systematic collection and decomposition of various data sources and abundant information regarding the pQSARs, including AADs, MLMs, data sets, peptide sequences, measured activities, model statistics, and literatures. The database also provides a comparison function for the various previously built pQSAR models reported by different groups via distinct approaches. The structured and searchable PepQSAR database is expected to provide a useful resource and powerful tool for the computational peptidology community, which is freely available at http://i.uestc.edu.cn/PQsarDB .
Collapse
Affiliation(s)
- Jing Lin
- Center for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC), No. 2006 Xiyuan Ave, West Hi-Tech Zone, Chengdu, 611731, China
| | - Li Wen
- Center for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC), No. 2006 Xiyuan Ave, West Hi-Tech Zone, Chengdu, 611731, China
| | - Yuwei Zhou
- Center for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC), No. 2006 Xiyuan Ave, West Hi-Tech Zone, Chengdu, 611731, China
| | - Shaozhou Wang
- Center for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC), No. 2006 Xiyuan Ave, West Hi-Tech Zone, Chengdu, 611731, China
| | - Haiyang Ye
- Center for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC), No. 2006 Xiyuan Ave, West Hi-Tech Zone, Chengdu, 611731, China
| | - Jun Su
- College of Music, Chengdu Normal University, Chengdu, 611130, China
| | - Juelin Li
- Center for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC), No. 2006 Xiyuan Ave, West Hi-Tech Zone, Chengdu, 611731, China
| | - Jianping Shu
- Center for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC), No. 2006 Xiyuan Ave, West Hi-Tech Zone, Chengdu, 611731, China
| | - Jian Huang
- Center for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC), No. 2006 Xiyuan Ave, West Hi-Tech Zone, Chengdu, 611731, China.
| | - Peng Zhou
- Center for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC), No. 2006 Xiyuan Ave, West Hi-Tech Zone, Chengdu, 611731, China.
| |
Collapse
|
4
|
Chou WC, Lin Z. Machine learning and artificial intelligence in physiologically based pharmacokinetic modeling. Toxicol Sci 2023; 191:1-14. [PMID: 36156156 PMCID: PMC9887681 DOI: 10.1093/toxsci/kfac101] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Physiologically based pharmacokinetic (PBPK) models are useful tools in drug development and risk assessment of environmental chemicals. PBPK model development requires the collection of species-specific physiological, and chemical-specific absorption, distribution, metabolism, and excretion (ADME) parameters, which can be a time-consuming and expensive process. This raises a need to create computational models capable of predicting input parameter values for PBPK models, especially for new compounds. In this review, we summarize an emerging paradigm for integrating PBPK modeling with machine learning (ML) or artificial intelligence (AI)-based computational methods. This paradigm includes 3 steps (1) obtain time-concentration PK data and/or ADME parameters from publicly available databases, (2) develop ML/AI-based approaches to predict ADME parameters, and (3) incorporate the ML/AI models into PBPK models to predict PK summary statistics (eg, area under the curve and maximum plasma concentration). We also discuss a neural network architecture "neural ordinary differential equation (Neural-ODE)" that is capable of providing better predictive capabilities than other ML methods when used to directly predict time-series PK profiles. In order to support applications of ML/AI methods for PBPK model development, several challenges should be addressed (1) as more data become available, it is important to expand the training set by including the structural diversity of compounds to improve the prediction accuracy of ML/AI models; (2) due to the black box nature of many ML models, lack of sufficient interpretability is a limitation; (3) Neural-ODE has great potential to be used to generate time-series PK profiles for new compounds with limited ADME information, but its application remains to be explored. Despite existing challenges, ML/AI approaches will continue to facilitate the efficient development of robust PBPK models for a large number of chemicals.
Collapse
Affiliation(s)
- Wei-Chun Chou
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32610, USA
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32608, USA
| | - Zhoumeng Lin
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32610, USA
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32608, USA
| |
Collapse
|
5
|
Li Q, Ma S, Zhang X, Zhai Z, Zhou L, Tao H, Wang Y, Pan J. DDPD 1.0: a manually curated and standardized database of digital properties of approved drugs for drug-likeness evaluation and drug development. Database (Oxford) 2022; 2022:6525313. [PMID: 35139189 PMCID: PMC9245338 DOI: 10.1093/database/baab083] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 10/23/2021] [Accepted: 01/06/2022] [Indexed: 11/22/2022]
Abstract
Drug-likeness is a vital consideration when selecting compounds in the early stage of
drug discovery. A series of drug-like properties are needed to predict the drug-likeness
of a given compound and provide useful guidelines to increase the likelihood of converting
lead compounds into drugs. Experimental physicochemical properties,
pharmacokinetic/toxicokinetic properties and maximum dosages of approved small-molecule
drugs from multiple text-based unstructured data resources have been manually assembled,
curated, further digitized and processed into structured data, which are deposited in the
Database of Digital Properties of approved Drugs (DDPD). DDPD 1.0 contains 30 212 drug
property entries, including 2250 approved drugs and 32 properties, in a standardized
value/unit format. Moreover, two analysis tools are provided to examine the drug-likeness
features of given molecules based on the collected property data of approved drugs.
Additionally, three case studies are presented to demonstrate how users can utilize the
database. We believe that this database will be a valuable resource for the drug discovery
and development field. Database URL: http://www.inbirg.com/ddpd
Collapse
Affiliation(s)
- Qiang Li
- Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District , Chongqing 400016, China
| | - Shiyong Ma
- Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District , Chongqing 400016, China
| | - Xuelu Zhang
- Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District , Chongqing 400016, China
| | - Zhaoyu Zhai
- Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District , Chongqing 400016, China
| | - Lu Zhou
- Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District , Chongqing 400016, China
| | - Haodong Tao
- Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District , Chongqing 400016, China
| | - Yachen Wang
- Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District , Chongqing 400016, China
| | - Jianbo Pan
- Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District , Chongqing 400016, China
| |
Collapse
|
6
|
Sohlenius-Sternbeck AK, Terelius Y. Evaluation of ADMET Predictor in Early Discovery Drug Metabolism and Pharmacokinetics Project Work. Drug Metab Dispos 2022; 50:95-104. [PMID: 34750195 DOI: 10.1124/dmd.121.000552] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 11/05/2021] [Indexed: 11/22/2022] Open
Abstract
A dataset consisting of measured values for LogD, solubility, metabolic stability in human liver microsomes (HLMs), and Caco-2 permeability was used to evaluate the prediction models for lipophilicity (S+LogD), water solubility (S+Sw_pH), metabolic stability in HLM (CYP_HLM_Clint), intestinal permeability (S+Peff), and P-glycoprotein (P-gp) substrate identification (P-gp substrate) in the software ADMET Predictor (AP) from Simulations Plus. The dataset consisted of a total of 4,794 compounds, with at least data from metabolic stability determinations in HLM, from multiple discovery projects at Medivir. Our evaluation shows that the global AP models can be used for categorization of high and low values based on predicted results for metabolic stability in HLM and intestinal permeability, and to give good predictions of LogD (R2= 0.79), guiding the synthesis of new compounds and for prioritizing in vitro ADME experiments. The model seems to overpredict solubility for the Medivir compounds, however. We also used the in-house datasets to build local models for LogD, solubility, metabolic stability, and permeability by using artificial neural network (ANN) models in the optional Modeler module of AP. Predictions of the test sets were performed with both the global and the local models, and the R2 values for linear regression for predicted versus measured HLM in vitro intrinsic clearance (CLint) based on logarithmic data were 0.72 for the in-house model and 0.53 for the AP model. The improved predictions with the local models are likely explained both by the specific chemical space of the Medivir dataset and laboratory-specific assay conditions for parameters that require biologic assay systems. SIGNIFICANCE STATEMENT: AP is useful early in projects for predicting and categorizing LogD, metabolic stability, and permeability, to guide the synthesis of new compounds, and for prioritizing in vitro ADME experiments. The building of local in-house prediction models with the optional AP Modeler Module can yield improved prediction success since these models are built on data from the same experimental setup and can also be based on compounds with similar structures.
Collapse
Affiliation(s)
- Anna-Karin Sohlenius-Sternbeck
- Research Institutes of Sweden, Södertälje, Sweden (A.-K.S.-S.); ADMEYT AB, Stenhamra, Sweden (Y.T.); and Medivir AB, Huddinge, Sweden (A.-K.S.-S., Y.T.)
| | - Ylva Terelius
- Research Institutes of Sweden, Södertälje, Sweden (A.-K.S.-S.); ADMEYT AB, Stenhamra, Sweden (Y.T.); and Medivir AB, Huddinge, Sweden (A.-K.S.-S., Y.T.)
| |
Collapse
|
7
|
Singh S, Singh DB, Gautam B, Singh A, Yadav N. Pharmacokinetics and pharmacodynamics analysis of drug candidates. Bioinformatics 2022. [DOI: 10.1016/b978-0-323-89775-4.00001-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
8
|
Danishuddin, Kumar V, Faheem M, Woo Lee K. A decade of machine learning-based predictive models for human pharmacokinetics: Advances and challenges. Drug Discov Today 2021; 27:529-537. [PMID: 34592448 DOI: 10.1016/j.drudis.2021.09.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/21/2021] [Accepted: 09/22/2021] [Indexed: 11/28/2022]
Abstract
Traditionally, in vitro and in vivo methods are useful for estimating human pharmacokinetics (PK) parameters; however, it is impractical to perform these complex and expensive experiments on a large number of compounds. The integration of publicly available chemical, or medical Big Data and artificial intelligence (AI)-based approaches led to qualitative and quantitative prediction of human PK of a candidate drug. However, predicting drug response with these approaches is challenging, partially because of the adaptation of algorithmic and limitations related to experimental data. In this report, we provide an overview of machine learning (ML)-based quantitative structure-activity relationship (QSAR) models used in the assessment or prediction of PK values as well as databases available for obtaining such data.
Collapse
Affiliation(s)
- Danishuddin
- Department of Bio & Medical Big Data (BK4), Division of Life Sciences, Research Institute of Natural Sciences (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Republic of Korea
| | - Vikas Kumar
- Department of Bio & Medical Big Data (BK4), Division of Life Sciences, Research Institute of Natural Sciences (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Republic of Korea
| | - Mohammad Faheem
- Department of Biotechnology, Indian Institute of Technology, Roorkee, Uttarakhand 247667, India
| | - Keun Woo Lee
- Department of Bio & Medical Big Data (BK4), Division of Life Sciences, Research Institute of Natural Sciences (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Republic of Korea.
| |
Collapse
|
9
|
Ouyang Y, Huang JJ, Wang YL, Zhong H, Song BA, Hao GF. In Silico Resources of Drug-Likeness as a Mirror: What Are We Lacking in Pesticide-Likeness? JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:10761-10773. [PMID: 34516106 DOI: 10.1021/acs.jafc.1c01460] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Unfavorable bioavailability is an important aspect underlying the failure of drug candidates. Computational approaches for evaluating drug-likeness can minimize these risks. Over the past decades, computational approaches for evaluating drug-likeness have sped up the process of drug development and were also quickly derived to pesticide-likeness. As a result of many critical differences between drugs and pesticides, many kinds of methods for drug-likeness cannot be used for pesticide-likeness. Therefore, it is crucial to comprehensively compare and analyze the differences between drug-likeness and pesticide-likeness, which may provide a basis for solving the problems encountered during the evaluation of pesticide-likeness. Here, we systematically collected the recent advances of drug-likeness and pesticide-likeness and compared their characteristics. We also evaluated the current lack of studies on pesticide-likeness, the molecular descriptors and parameters adopted, the pesticide-likeness model on pesticide target organisms, and comprehensive analysis tools. This work may guide researchers to use appropriate methods for developing pesticide-likeness models. It may also aid non-specialists to understand some important concepts in drug-likeness and pesticide-likeness.
Collapse
Affiliation(s)
- Yan Ouyang
- Guizhou Engineering Laboratory for Synthetic Drugs, Key Laboratory of Guizhou Fermentation Engineering and Biomedicine, School of Pharmaceutical Sciences, Guizhou University, Guiyang, Guizhou 550025, People's Republic of China
| | - Jun-Jie Huang
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Research and Development Center for Fine Chemicals, Guizhou University, Guiyang, Guizhou 550025, People's Republic of China
| | - Yu-Liang Wang
- Key Laboratory of Pesticide & Chemical Biology, Ministry of Education, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, People's Republic of China
- International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan, Hubei 430079, People's Republic of China
| | - Hang Zhong
- Guizhou Engineering Laboratory for Synthetic Drugs, Key Laboratory of Guizhou Fermentation Engineering and Biomedicine, School of Pharmaceutical Sciences, Guizhou University, Guiyang, Guizhou 550025, People's Republic of China
| | - Bao-An Song
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Research and Development Center for Fine Chemicals, Guizhou University, Guiyang, Guizhou 550025, People's Republic of China
| | - Ge-Fei Hao
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Research and Development Center for Fine Chemicals, Guizhou University, Guiyang, Guizhou 550025, People's Republic of China
| |
Collapse
|
10
|
Evaluation of Quantitative Structure Property Relationship Algorithms for Predicting Plasma Protein Binding in Humans. ACTA ACUST UNITED AC 2021; 17:100142. [PMID: 34017929 DOI: 10.1016/j.comtox.2020.100142] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The extent of plasma protein binding is an important compound-specific property that influences a compound's pharmacokinetic behavior and is a critical input parameter for predicting exposure in physiologically based pharmacokinetic (PBPK) modeling. When experimentally determined fraction unbound in plasma (fup) data are not available, quantitative structure-property relationship (QSPR) models can be used for prediction. Because available QSPR models were developed based on training sets containing pharmaceutical-like compounds, we compared their prediction accuracy for environmentally relevant and pharmaceutical compounds. Fup values were calculated using Ingle et al., Watanabe et al. and ADMET Predictor (Simulation Plus). The test set included 818 pharmaceutical and environmentally relevant compounds with fup values ranging from 0.01 to 1. Overall, the three QSPR models resulted in over-prediction of fup for highly binding compounds and under-prediction for low or moderately binding compounds. For highly binding compounds (0.01≤ fup ≤ 0.25), Watanabe et al. performed better with a lower mean absolute error (MAE) of 6.7% and a lower mean absolute relative prediction error (RPE) of 171.7 % than other methods. For low to moderately binding compounds, both Ingle et al. and ADMET Predictor performed better than Watanabe et al. with superior MAE and RPE values. The positive polar surface area, the number of basic functional groups and lipophilicity were the most important chemical descriptors for predicting fup. This study demonstrated that the prediction of fup was the most uncertain for highly binding compounds. This suggested that QSPR-predicted fup values should be used with caution in PBPK modeling.
Collapse
|
11
|
Grzegorzewski J, Brandhorst J, Green K, Eleftheriadou D, Duport Y, Barthorscht F, Köller A, Ke DYJ, De Angelis S, König M. PK-DB: pharmacokinetics database for individualized and stratified computational modeling. Nucleic Acids Res 2021; 49:D1358-D1364. [PMID: 33151297 PMCID: PMC7779054 DOI: 10.1093/nar/gkaa990] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 10/01/2020] [Accepted: 11/03/2020] [Indexed: 11/13/2022] Open
Abstract
A multitude of pharmacokinetics studies have been published. However, due to the lack of an open database, pharmacokinetics data, as well as the corresponding meta-information, have been difficult to access. We present PK-DB (https://pk-db.com), an open database for pharmacokinetics information from clinical trials. PK-DB provides curated information on (i) characteristics of studied patient cohorts and subjects (e.g. age, bodyweight, smoking status, genetic variants); (ii) applied interventions (e.g. dosing, substance, route of application); (iii) pharmacokinetic parameters (e.g. clearance, half-life, area under the curve) and (iv) measured pharmacokinetic time-courses. Key features are the representation of experimental errors, the normalization of measurement units, annotation of information to biological ontologies, calculation of pharmacokinetic parameters from concentration-time profiles, a workflow for collaborative data curation, strong validation rules on the data, computational access via a REST API as well as human access via a web interface. PK-DB enables meta-analysis based on data from multiple studies and data integration with computational models. A special focus lies on meta-data relevant for individualized and stratified computational modeling with methods like physiologically based pharmacokinetic (PBPK), pharmacokinetic/pharmacodynamic (PK/PD), or population pharmacokinetic (pop PK) modeling.
Collapse
Affiliation(s)
- Jan Grzegorzewski
- Institute for Theoretical Biology, Humboldt-University Berlin, Invalidenstraße 110, Berlin 10115, Germany
| | - Janosch Brandhorst
- Institute for Theoretical Biology, Humboldt-University Berlin, Invalidenstraße 110, Berlin 10115, Germany
| | - Kathleen Green
- Department of Biochemistry, University of Stellenbosch, Van der Byl Street, Stellenbosch 7600, South Africa
| | - Dimitra Eleftheriadou
- Institute for Theoretical Biology, Humboldt-University Berlin, Invalidenstraße 110, Berlin 10115, Germany
| | - Yannick Duport
- Department of Mathematics and Computer Science, Freie Universität Berlin, Arnimallee 14, Berlin 14195, Germany
| | - Florian Barthorscht
- Institute for Theoretical Biology, Humboldt-University Berlin, Invalidenstraße 110, Berlin 10115, Germany
| | - Adrian Köller
- Institute for Theoretical Biology, Humboldt-University Berlin, Invalidenstraße 110, Berlin 10115, Germany
| | - Danny Yu Jia Ke
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Sara De Angelis
- King's College London, Department of Biomedical Engineering & Imaging Sciences, London, UK
| | - Matthias König
- Institute for Theoretical Biology, Humboldt-University Berlin, Invalidenstraße 110, Berlin 10115, Germany
| |
Collapse
|
12
|
Gonzalez Hernandez F, Carter SJ, Iso-Sipilä J, Goldsmith P, Almousa AA, Gastine S, Lilaonitkul W, Kloprogge F, Standing JF. An automated approach to identify scientific publications reporting pharmacokinetic parameters. Wellcome Open Res 2021; 6:88. [PMID: 34381873 PMCID: PMC8343403 DOI: 10.12688/wellcomeopenres.16718.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2021] [Indexed: 11/29/2022] Open
Abstract
Pharmacokinetic (PK) predictions of new chemical entities are aided by prior knowledge from other compounds. The development of robust algorithms that improve preclinical and clinical phases of drug development remains constrained by the need to search, curate and standardise PK information across the constantly-growing scientific literature. The lack of centralised, up-to-date and comprehensive repositories of PK data represents a significant limitation in the drug development pipeline.In this work, we propose a machine learning approach to automatically identify and characterise scientific publications reporting PK parameters from in vivo data, providing a centralised repository of PK literature. A dataset of 4,792 PubMed publications was labelled by field experts depending on whether in vivo PK parameters were estimated in the study. Different classification pipelines were compared using a bootstrap approach and the best-performing architecture was used to develop a comprehensive and automatically-updated repository of PK publications. The best-performing architecture encoded documents using unigram features and mean pooling of BioBERT embeddings obtaining an F1 score of 83.8% on the test set. The pipeline retrieved over 121K PubMed publications in which in vivo PK parameters were estimated and it was scheduled to perform weekly updates on newly published articles. All the relevant documents were released through a publicly available web interface (https://app.pkpdai.com) and characterised by the drugs, species and conditions mentioned in the abstract, to facilitate the subsequent search of relevant PK data. This automated, open-access repository can be used to accelerate the search and comparison of PK results, curate ADME datasets, and facilitate subsequent text mining tasks in the PK domain.
Collapse
Affiliation(s)
| | - Simon J Carter
- Institute of Pharmacy, Uppsala University, Uppsala, Sweden.,Institute for Global Health, University College London, London, UK
| | | | | | | | - Silke Gastine
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Watjana Lilaonitkul
- Institute of Health Informatics, University College London, London, UK.,Health Data Research, London, UK
| | - Frank Kloprogge
- Institute for Global Health, University College London, London, UK
| | - Joseph F Standing
- Great Ormond Street Institute of Child Health, University College London, London, UK
| |
Collapse
|
13
|
Biological evaluation and pharmacokinetic profiling of a coumarin-benzothiazole hybrid as a new scaffold for human gliomas. MEDICINE IN DRUG DISCOVERY 2020. [DOI: 10.1016/j.medidd.2020.100012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
14
|
Zafar F, Gupta A, Thangavel K, Khatana K, Sani AA, Ghosal A, Tandon P, Nishat N. Physicochemical and Pharmacokinetic Analysis of Anacardic Acid Derivatives. ACS OMEGA 2020; 5:6021-6030. [PMID: 32226883 PMCID: PMC7098041 DOI: 10.1021/acsomega.9b04398] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 02/18/2020] [Indexed: 05/23/2023]
Abstract
Anacardic acid (AA) and its derivatives are well-known for their therapeutic applications ranging from antitumor, antibacterial, antioxidant, anticancer, and so forth. However, their poor pharmacokinetic and safety properties create significant hurdles in the formulation of the final drug molecule. As a part of our endeavor to enhance the potential and exploration of the anticancer activities, a detailed study on the properties of selected AA derivatives was performed in this work. A comprehensive analysis of the drug-like properties of 100 naturally occurring AA derivatives was performed, and the results were compared with certain marketed anticancer drugs. The work focused on the understanding of the interplay among eight physicochemical properties. The relationships between the physicochemical properties, absorption, distribution, metabolism, and excretion attributes, and the in silico toxicity profile for the set of AA derivatives were established. The ligand efficacy of the finally scrutinized 17 AA derivatives on the basis of pharmacokinetic properties and toxicity parameters was further subjected to dock against the potential anticancer target cyclin-dependent kinase 2 (PDB ID: 1W98). In the docked complex, the ligand molecules (AA derivatives) selectively bind with the target residues, and a high binding affinity of the ligand molecules was ensured by the full fitness score using the SwissDock Web server. The BOILED-Egg model shows that out of 17 scrutinized molecules, 3 molecules exhibit gastrointestinal absorption capability and 14 molecules exhibit permeability through the blood-brain barrier penetration. The analysis can also provide some useful insights to chemists to modify the existing natural scaffolds in designing new anacardic anticancer drugs. The increased probability of success may lead to the identification of drug-like candidates with favorable safety profiles after further clinical evaluation.
Collapse
Affiliation(s)
- Fahmina Zafar
- Inorganic
Materials Research Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, India
| | - Anjali Gupta
- Division
of Chemistry, School of Basic and Applied Science, Galgotias University, Greater
Noida 201310, Uttar Pradesh, India
| | - Karthick Thangavel
- Department
of Physics, School of Electrical and Electronics Engineering, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India
| | - Kavita Khatana
- Division
of Chemistry, School of Basic and Applied Science, Galgotias University, Greater
Noida 201310, Uttar Pradesh, India
| | - Ali Alhaji Sani
- Division
of Chemistry, School of Basic and Applied Science, Galgotias University, Greater
Noida 201310, Uttar Pradesh, India
| | - Anujit Ghosal
- Inorganic
Materials Research Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, India
- Division
of Chemistry, School of Basic and Applied Science, Galgotias University, Greater
Noida 201310, Uttar Pradesh, India
- School
of Life Sciences, Beijing Institute of Technology, Beijing 100811, China
| | - Poonam Tandon
- Department
of Physics, University of Lucknow, Lucknow 226007, India
| | - Nahid Nishat
- Inorganic
Materials Research Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, India
| |
Collapse
|
15
|
Yang X, Wang Y, Byrne R, Schneider G, Yang S. Concepts of Artificial Intelligence for Computer-Assisted Drug Discovery. Chem Rev 2019; 119:10520-10594. [PMID: 31294972 DOI: 10.1021/acs.chemrev.8b00728] [Citation(s) in RCA: 421] [Impact Index Per Article: 70.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Artificial intelligence (AI), and, in particular, deep learning as a subcategory of AI, provides opportunities for the discovery and development of innovative drugs. Various machine learning approaches have recently (re)emerged, some of which may be considered instances of domain-specific AI which have been successfully employed for drug discovery and design. This review provides a comprehensive portrayal of these machine learning techniques and of their applications in medicinal chemistry. After introducing the basic principles, alongside some application notes, of the various machine learning algorithms, the current state-of-the art of AI-assisted pharmaceutical discovery is discussed, including applications in structure- and ligand-based virtual screening, de novo drug design, physicochemical and pharmacokinetic property prediction, drug repurposing, and related aspects. Finally, several challenges and limitations of the current methods are summarized, with a view to potential future directions for AI-assisted drug discovery and design.
Collapse
Affiliation(s)
- Xin Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital , Sichuan University , Chengdu , Sichuan 610041 , China
| | - Yifei Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital , Sichuan University , Chengdu , Sichuan 610041 , China
| | - Ryan Byrne
- ETH Zurich , Department of Chemistry and Applied Biosciences , Vladimir-Prelog-Weg 4 , CH-8093 Zurich , Switzerland
| | - Gisbert Schneider
- ETH Zurich , Department of Chemistry and Applied Biosciences , Vladimir-Prelog-Weg 4 , CH-8093 Zurich , Switzerland
| | - Shengyong Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital , Sichuan University , Chengdu , Sichuan 610041 , China
| |
Collapse
|
16
|
Abstract
Drug discovery has evolved significantly over the past two decades. Progress in key areas such as molecular and structural biology has contributed to the elucidation of the three-dimensional structure and function of a wide range of biological molecules of therapeutic interest. In this context, the integration of experimental techniques, such as X-ray crystallography, and computational methods, such as molecular docking, has promoted the emergence of several areas in drug discovery, such as structure-based drug design (SBDD). SBDD strategies have been broadly used to identify, predict and optimize the activity of small molecules toward a molecular target and have contributed to major scientific breakthroughs in pharmaceutical R&D. This chapter outlines molecular docking and structure-based virtual screening (SBVS) protocols used to predict the interaction of small molecules with the phosphatidylinositol-bisphosphate-kinase PI3Kδ, which is a molecular target for hematological diseases. A detailed description of the molecular docking and SBVS procedures and an evaluation of the results are provided.
Collapse
|
17
|
Cabrera-Pérez MÁ, Pham-The H. Computational modeling of human oral bioavailability: what will be next? Expert Opin Drug Discov 2018; 13:509-521. [PMID: 29663836 DOI: 10.1080/17460441.2018.1463988] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION The oral route is the most convenient way of administrating drugs. Therefore, accurate determination of oral bioavailability is paramount during drug discovery and development. Quantitative structure-property relationship (QSPR), rule-of-thumb (RoT) and physiologically based-pharmacokinetic (PBPK) approaches are promising alternatives to the early oral bioavailability prediction. Areas covered: The authors give insight into the factors affecting bioavailability, the fundamental theoretical framework and the practical aspects of computational methods for predicting this property. They also give their perspectives on future computational models for estimating oral bioavailability. Expert opinion: Oral bioavailability is a multi-factorial pharmacokinetic property with its accurate prediction challenging. For RoT and QSPR modeling, the reliability of datasets, the significance of molecular descriptor families and the diversity of chemometric tools used are important factors that define model predictability and interpretability. Likewise, for PBPK modeling the integrity of the pharmacokinetic data, the number of input parameters, the complexity of statistical analysis and the software packages used are relevant factors in bioavailability prediction. Although these approaches have been utilized independently, the tendency to use hybrid QSPR-PBPK approaches together with the exploration of ensemble and deep-learning systems for QSPR modeling of oral bioavailability has opened new avenues for development promising tools for oral bioavailability prediction.
Collapse
Affiliation(s)
- Miguel Ángel Cabrera-Pérez
- a Unit of Modeling and Experimental Biopharmaceutics , Chemical Bioactive Center, Central University of Las Villas , Santa Clara , Cuba.,b Department of Pharmacy and Pharmaceutical Technology , University of Valencia , Burjassot , Spain.,c Department of Engineering, Area of Pharmacy and Pharmaceutical Technology , Miguel Hernández University , Alicante , Spain
| | - Hai Pham-The
- d Department of Pharmaceutical Chemistry , Hanoi University of Pharmacy , Hanoi , Vietnam
| |
Collapse
|
18
|
Douguet D. Data Sets Representative of the Structures and Experimental Properties of FDA-Approved Drugs. ACS Med Chem Lett 2018. [PMID: 29541361 DOI: 10.1021/acsmedchemlett.7b00462] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Presented here are several data sets that gather information collected from the labels of the FDA approved drugs: their molecular structures and those of the described active metabolites, their associated pharmacokinetics and pharmacodynamics data, and the history of their marketing authorization by the FDA. To date, 1852 chemical structures have been identified with a molecular weight less than 2000 of which 492 are or have active metabolites. To promote the sharing of data, the original web server was upgraded for browsing the database and downloading the data sets (http://chemoinfo.ipmc.cnrs.fr/edrug3d). It is believed that the multidimensional chemistry-oriented collections are an essential resource for a thorough analysis of the current drug chemical space. The data sets are envisioned as being used in a wide range of endeavors that include drug repurposing, drug design, privileged structures analyses, structure-activity relationship studies, and improving of absorption, distribution, metabolism, and elimination predictive models.
Collapse
Affiliation(s)
- Dominique Douguet
- Université Côte d’Azur, Inserm, CNRS, IPMC, 660 Route des Lucioles, 06560 Valbonne, France
| |
Collapse
|
19
|
FERREIRA LEONARDOG, OLIVA GLAUCIUS, ANDRICOPULO ADRIANOD. From Medicinal Chemistry to Human Health: Current Approaches to Drug Discovery for Cancer and Neglected Tropical Diseases. ACTA ACUST UNITED AC 2018; 90:645-661. [DOI: 10.1590/0001-3765201820170505] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 08/09/2017] [Indexed: 12/18/2022]
|
20
|
Zheng M, Zhao J, Cui C, Fu Z, Li X, Liu X, Ding X, Tan X, Li F, Luo X, Chen K, Jiang H. Computational chemical biology and drug design: Facilitating protein structure, function, and modulation studies. Med Res Rev 2018; 38:914-950. [DOI: 10.1002/med.21483] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/13/2017] [Accepted: 12/15/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Mingyue Zheng
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
| | - Jihui Zhao
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
| | - Chen Cui
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
| | - Zunyun Fu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
| | - Xutong Li
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
| | - Xiaohong Liu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
- School of Life Science and Technology; ShanghaiTech University; Shanghai China
| | - Xiaoyu Ding
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
| | - Xiaoqin Tan
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
| | - Fei Li
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
- Department of Chemistry, College of Sciences; Shanghai University; Shanghai China
| | - Xiaomin Luo
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
| | - Kaixian Chen
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
- School of Life Science and Technology; ShanghaiTech University; Shanghai China
| | - Hualiang Jiang
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
| |
Collapse
|
21
|
Sun L, Yang H, Li J, Wang T, Li W, Liu G, Tang Y. In Silico Prediction of Compounds Binding to Human Plasma Proteins by QSAR Models. ChemMedChem 2017; 13:572-581. [DOI: 10.1002/cmdc.201700582] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 10/18/2017] [Indexed: 12/18/2022]
Affiliation(s)
- Lixia Sun
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy; East China University of Science and Technology; Shanghai 200237 China
| | - Hongbin Yang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy; East China University of Science and Technology; Shanghai 200237 China
| | - Jie Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy; East China University of Science and Technology; Shanghai 200237 China
| | - Tianduanyi Wang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy; East China University of Science and Technology; Shanghai 200237 China
| | - Weihua Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy; East China University of Science and Technology; Shanghai 200237 China
| | - Guixia Liu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy; East China University of Science and Technology; Shanghai 200237 China
| | - Yun Tang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy; East China University of Science and Technology; Shanghai 200237 China
| |
Collapse
|
22
|
Pilon AC, Valli M, Dametto AC, Pinto MEF, Freire RT, Castro-Gamboa I, Andricopulo AD, Bolzani VS. NuBBE DB: an updated database to uncover chemical and biological information from Brazilian biodiversity. Sci Rep 2017; 7:7215. [PMID: 28775335 PMCID: PMC5543130 DOI: 10.1038/s41598-017-07451-x] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/28/2017] [Indexed: 01/24/2023] Open
Abstract
The intrinsic value of biodiversity extends beyond species diversity, genetic heritage, ecosystem variability and ecological services, such as climate regulation, water quality, nutrient cycling and the provision of reproductive habitats it is also an inexhaustible source of molecules and products beneficial to human well-being. To uncover the chemistry of Brazilian natural products, the Nuclei of Bioassays, Ecophysiology and Biosynthesis of Natural Products Database (NuBBEDB) was created as the first natural product library from Brazilian biodiversity. Since its launch in 2013, the NuBBEDB has proven to be an important resource for new drug design and dereplication studies. Consequently, continuous efforts have been made to expand its contents and include a greater diversity of natural sources to establish it as a comprehensive compendium of available biogeochemical information about Brazilian biodiversity. The content in the NuBBEDB is freely accessible online (https://nubbe.iq.unesp.br/portal/nubbedb.html) and provides validated multidisciplinary information, chemical descriptors, species sources, geographic locations, spectroscopic data (NMR) and pharmacological properties. Herein, we report the latest advancements concerning the interface, content and functionality of the NuBBEDB. We also present a preliminary study on the current profile of the compounds present in Brazilian territory.
Collapse
Affiliation(s)
- Alan C Pilon
- Nuclei of Bioassays, Biosynthesis and Ecophysiology of Natural Products (NuBBE), Department of Organic Chemistry, Institute of Chemistry, Sao Paulo State University - UNESP, 14800-060, Araraquara, SP, Brazil
| | - Marilia Valli
- Nuclei of Bioassays, Biosynthesis and Ecophysiology of Natural Products (NuBBE), Department of Organic Chemistry, Institute of Chemistry, Sao Paulo State University - UNESP, 14800-060, Araraquara, SP, Brazil
| | - Alessandra C Dametto
- Nuclei of Bioassays, Biosynthesis and Ecophysiology of Natural Products (NuBBE), Department of Organic Chemistry, Institute of Chemistry, Sao Paulo State University - UNESP, 14800-060, Araraquara, SP, Brazil
| | - Meri Emili F Pinto
- Nuclei of Bioassays, Biosynthesis and Ecophysiology of Natural Products (NuBBE), Department of Organic Chemistry, Institute of Chemistry, Sao Paulo State University - UNESP, 14800-060, Araraquara, SP, Brazil
| | - Rafael T Freire
- Centro de Imagens e Espectroscopia in vivo por Ressonância Magnética, Institute of Physics of Sao Carlos, University of Sao Paulo - USP, 13566-590, Sao Carlos, SP, Brazil
| | - Ian Castro-Gamboa
- Nuclei of Bioassays, Biosynthesis and Ecophysiology of Natural Products (NuBBE), Department of Organic Chemistry, Institute of Chemistry, Sao Paulo State University - UNESP, 14800-060, Araraquara, SP, Brazil
| | - Adriano D Andricopulo
- Laboratório de Química Medicinal e Computacional (LQMC), Centro de Pesquisa e Inovação em Biodiversidade eFármacos, Institute of Physics of Sao Carlos, University of Sao Paulo - USP, 13563-120, Sao Carlos, SP, Brazil
| | - Vanderlan S Bolzani
- Nuclei of Bioassays, Biosynthesis and Ecophysiology of Natural Products (NuBBE), Department of Organic Chemistry, Institute of Chemistry, Sao Paulo State University - UNESP, 14800-060, Araraquara, SP, Brazil.
| |
Collapse
|
23
|
Xu Q, Liu K, Lin X, Qin Y, Chen L, Cheng J, Zhong M, He Q, Li Y, Wang T, Pan J, Peng M, Yao L, Ji Z. ADMETNet: The knowledge base of pharmacokinetics and toxicology network. J Genet Genomics 2017; 44:273-276. [PMID: 28529076 DOI: 10.1016/j.jgg.2017.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/11/2017] [Accepted: 04/29/2017] [Indexed: 10/19/2022]
Affiliation(s)
- Quan Xu
- State Key Laboratory of Stress Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Ke Liu
- State Key Laboratory of Stress Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Xingming Lin
- State Key Laboratory of Stress Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Yangmei Qin
- State Key Laboratory of Stress Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Linshan Chen
- State Key Laboratory of Stress Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Jiao Cheng
- State Key Laboratory of Stress Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Mindong Zhong
- State Key Laboratory of Stress Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Qiushun He
- State Key Laboratory of Stress Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Yinbo Li
- State Key Laboratory of Stress Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Tingwu Wang
- State Key Laboratory of Stress Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Jianbo Pan
- State Key Laboratory of Stress Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Menglu Peng
- State Key Laboratory of Stress Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Lixia Yao
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Zhiliang Ji
- State Key Laboratory of Stress Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China; The Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, Xiamen 361005, China.
| |
Collapse
|
24
|
Przybylak K, Madden J, Covey-Crump E, Gibson L, Barber C, Patel M, Cronin M. Characterisation of data resources for in silico modelling: benchmark datasets for ADME properties. Expert Opin Drug Metab Toxicol 2017; 14:169-181. [DOI: 10.1080/17425255.2017.1316449] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- K.R. Przybylak
- School of Pharmacy and Chemistry, Liverpool John Moores University, Liverpool, UK
| | - J.C. Madden
- School of Pharmacy and Chemistry, Liverpool John Moores University, Liverpool, UK
| | | | - L. Gibson
- Lhasa Limited, Granary Wharf House, Leeds, UK
| | - C. Barber
- Lhasa Limited, Granary Wharf House, Leeds, UK
| | - M. Patel
- Lhasa Limited, Granary Wharf House, Leeds, UK
| | - M.T.D. Cronin
- School of Pharmacy and Chemistry, Liverpool John Moores University, Liverpool, UK
| |
Collapse
|
25
|
Legehar A, Xhaard H, Ghemtio L. IDAAPM: integrated database of ADMET and adverse effects of predictive modeling based on FDA approved drug data. J Cheminform 2016; 8:33. [PMID: 27303447 PMCID: PMC4906584 DOI: 10.1186/s13321-016-0141-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/20/2016] [Indexed: 02/05/2023] Open
Abstract
Background The disposition of a pharmaceutical compound within an organism, i.e. its Absorption, Distribution, Metabolism, Excretion, Toxicity (ADMET) properties and adverse effects, critically affects late stage failure of drug candidates and has led to the withdrawal of approved drugs. Computational methods are effective approaches to reduce the number of safety issues by analyzing possible links between chemical structures and ADMET or adverse effects, but this is limited by the size, quality, and heterogeneity of the data available from individual sources. Thus, large, clean and integrated databases of approved drug data, associated with fast and efficient predictive tools are desirable early in the drug discovery process. Description We have built a relational database (IDAAPM) to integrate available approved drug data such as drug approval information, ADMET and adverse effects, chemical structures and molecular descriptors, targets, bioactivity and related references. The database has been coupled with a searchable web interface and modern data analytics platform (KNIME) to allow data access, data transformation, initial analysis and further predictive modeling. Data were extracted from FDA resources and supplemented from other publicly available databases. Currently, the database contains information regarding about 19,226 FDA approval applications for 31,815 products (small molecules and biologics) with their approval history, 2505 active ingredients, together with as many ADMET properties, 1629 molecular structures, 2.5 million adverse effects and 36,963 experimental drug-target bioactivity data. Conclusion IDAAPM is a unique resource that, in a single relational database, provides detailed information on FDA approved drugs including their ADMET properties and adverse effects, the corresponding targets with bioactivity data, coupled with a data analytics platform. It can be used to perform basic to complex drug-target ADMET or adverse effects analysis and predictive modeling. IDAAPM is freely accessible at http://idaapm.helsinki.fi and can be exploited through a KNIME workflow connected to the database.FDA approved drug data integration for predictive modeling ![]() Electronic supplementary material The online version of this article (doi:10.1186/s13321-016-0141-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ashenafi Legehar
- Centre for Drug Research, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland
| | - Henri Xhaard
- Centre for Drug Research, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland ; Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Leo Ghemtio
- Centre for Drug Research, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland
| |
Collapse
|
26
|
LaLone CA, Berninger JP, Villeneuve DL, Ankley GT. Leveraging existing data for prioritization of the ecological risks of human and veterinary pharmaceuticals to aquatic organisms. Philos Trans R Soc Lond B Biol Sci 2015; 369:rstb.2014.0022. [PMID: 25405975 DOI: 10.1098/rstb.2014.0022] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Medicinal innovation has led to the discovery and use of thousands of human and veterinary drugs. With this comes the potential for unintended effects on non-target organisms exposed to pharmaceuticals inevitably entering the environment. The impracticality of generating whole-organism chronic toxicity data representative of all species in the environment has necessitated prioritization of drugs for focused empirical testing as well as field monitoring. Current prioritization strategies typically emphasize likelihood for exposure (i.e. predicted/measured environmental concentrations), while incorporating only rather limited consideration of potential effects of the drug to non-target organisms. However, substantial mammalian pharmacokinetic and mechanism/mode of action (MOA) data are produced during drug development to understand drug target specificity and efficacy for intended consumers. An integrated prioritization strategy for assessing risks of human and veterinary drugs would leverage available pharmacokinetic and toxicokinetic data for evaluation of the potential for adverse effects to non-target organisms. In this reiview, we demonstrate the utility of read-across approaches to leverage mammalian absorption, distribution, metabolism and elimination data; analyse cross-species molecular target conservation and translate therapeutic MOA to an adverse outcome pathway(s) relevant to aquatic organisms as a means to inform prioritization of drugs for focused toxicity testing and environmental monitoring.
Collapse
Affiliation(s)
- Carlie A LaLone
- Water Resources Center, College of Food, Agricultural and Natural Resource Sciences, University of Minnesota, 1985 Buford Avenue, St Paul, MN 55108, USA Office of Research and Development, National Health and Environmental Effects Research Laboratory, Mid-Continent Ecology Division, US Environmental Protection Agency, 6201 Congdon Boulevard, Duluth, MN 55804, USA
| | - Jason P Berninger
- National Research Council, 6201 Congdon Boulevard, Duluth, MN 55804, USA
| | - Daniel L Villeneuve
- Office of Research and Development, National Health and Environmental Effects Research Laboratory, Mid-Continent Ecology Division, US Environmental Protection Agency, 6201 Congdon Boulevard, Duluth, MN 55804, USA
| | - Gerald T Ankley
- Office of Research and Development, National Health and Environmental Effects Research Laboratory, Mid-Continent Ecology Division, US Environmental Protection Agency, 6201 Congdon Boulevard, Duluth, MN 55804, USA
| |
Collapse
|
27
|
Abdel-Aziz AAM, El-Azab AS, Alanazi AM, Asiri YA, Al-Suwaidan IA, Maarouf AR, Ayyad RR, Shawer TZ. Synthesis and potential antitumor activity of 7-(4-substituted piperazin-1-yl)-4-oxoquinolines based on ciprofloxacin and norfloxacin scaffolds: in silico studies. J Enzyme Inhib Med Chem 2015. [PMID: 26226179 DOI: 10.3109/14756366.2015.1069288] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The potential antitumor activities of a series of 7-(4-substituted piperazin-1-yl)fluoroquinolone derivatives (1-14a,b) using ciprofloxacin and norfloxacin as scaffolds are described. These compounds exhibit potent and broad spectrum antitumor activities using 60 human cell lines in addition to the inherent antibacterial activity. Compounds 1a, 2a, 3b, 6b and 7a were found to be the most potent, while 2b, 5b, and 6a were found to have an average activity. The results of this study demonstrated that compounds 1a, 2a, 3b, 6b and 7a (mean GI50; 2.63-3.09 µM) are nearly 7-fold more potent compared with the positive control 5-fluorouracil (mean GI50; 22.60 µM). More interestingly, compounds 1a, 2a, 3b, 6b and 7a have an almost antitumor activity similar to gefitinib (mean GI50; 3.24 µM) and are nearly 2-fold more potent compared to erlotinib (mean GI50; 7.29 µM). In silico study and ADME-Tox prediction methodology were used to study the antitumor activity of the most active compounds and to identify the structural features required for antitumor activity.
Collapse
Affiliation(s)
- Alaa A-M Abdel-Aziz
- a Department of Pharmaceutical Chemistry , College of Pharmacy, King Saud University , Riyadh , Saudi Arabia .,b Department of Medicinal Chemistry, Faculty of Pharmacy , University of Mansoura , Mansoura , Egypt
| | - Adel S El-Azab
- a Department of Pharmaceutical Chemistry , College of Pharmacy, King Saud University , Riyadh , Saudi Arabia .,c Department of Organic Chemistry, Faculty of Pharmacy , Al-Azhar University , Cairo , Egypt
| | - Amer M Alanazi
- a Department of Pharmaceutical Chemistry , College of Pharmacy, King Saud University , Riyadh , Saudi Arabia
| | - Yousif A Asiri
- d Department of Clinical Pharmacy , College of Pharmacy, King Saud University , Riyadh , Saudi Arabia
| | - Ibrahim A Al-Suwaidan
- a Department of Pharmaceutical Chemistry , College of Pharmacy, King Saud University , Riyadh , Saudi Arabia
| | - Azza R Maarouf
- b Department of Medicinal Chemistry, Faculty of Pharmacy , University of Mansoura , Mansoura , Egypt .,e Department of Pharmaceutical Chemistry, Faculty of Pharmacy , Delta University for Science & Technology , Gamasa City , Egypt , and
| | - Rezk R Ayyad
- f Department of Pharmaceutical Chemistry, Faculty of Pharmacy , Al-Azhar University , Cairo , Egypt
| | - Taghreed Z Shawer
- f Department of Pharmaceutical Chemistry, Faculty of Pharmacy , Al-Azhar University , Cairo , Egypt
| |
Collapse
|
28
|
Alanazi AM, Abdel-Aziz AAM, Shawer TZ, Ayyad RR, Al-Obaid AM, Al-Agamy MHM, Maarouf AR, El-Azab AS. Synthesis, antitumor and antimicrobial activity of some new 6-methyl-3-phenyl-4(3H)-quinazolinone analogues: in silico studies. J Enzyme Inhib Med Chem 2015; 31:721-35. [PMID: 26162029 DOI: 10.3109/14756366.2015.1060482] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Some new derivatives of substituted-4(3H)-quinazolinones were synthesized and evaluated for their in vitro antitumor and antimicrobial activities. The results of this study demonstrated that compound 5 yielded selective activities toward NSC Lung Cancer EKVX cell line, Colon Cancer HCT-15 cell line and Breast Cancer MDA-MB-231/ATCC cell line, while NSC Lung Cancer EKVX cell line and CNS Cancer SF-295 cell line were sensitive to compound 8. Additionally, compounds 12 and 13 showed moderate effectiveness toward numerous cell lines belonging to different tumor subpanels. On the other hand, the results of antimicrobial screening revealed that compounds 1, 9 and 14 are the most active against Staphylococcus aureus ATCC 29213 with minimum inhibitory concentration (MIC) of 16, 32 and 32 μg/mL respectively, while compound 14 possessed antimicrobial activities against all tested strains with the lowest MIC compared with other tested compounds. In silico study, ADME-Tox prediction and molecular docking methodology were used to study the antitumor activity and to identify the structural features required for antitumor activity.
Collapse
Affiliation(s)
- Amer M Alanazi
- a Department of Pharmaceutical Chemistry , College of Pharmacy, King Saud University , Riyadh , Saudi Arabia
| | - Alaa A-M Abdel-Aziz
- a Department of Pharmaceutical Chemistry , College of Pharmacy, King Saud University , Riyadh , Saudi Arabia .,b Department of Medicinal Chemistry , Faculty of Pharmacy, University of Mansoura , Mansoura , Egypt
| | - Taghreed Z Shawer
- c Department of Pharmaceutical Chemistry , Faculty of Pharmacy, Al-Azhar University , Cairo , Egypt
| | - Rezk R Ayyad
- c Department of Pharmaceutical Chemistry , Faculty of Pharmacy, Al-Azhar University , Cairo , Egypt
| | - Abdulrahman M Al-Obaid
- a Department of Pharmaceutical Chemistry , College of Pharmacy, King Saud University , Riyadh , Saudi Arabia
| | - Mohamed H M Al-Agamy
- d Department of Pharmaceutics and Microbiology , College of Pharmacy, King Saud University , Riyadh , Saudi Arabia , and
| | - Azza R Maarouf
- b Department of Medicinal Chemistry , Faculty of Pharmacy, University of Mansoura , Mansoura , Egypt
| | - Adel S El-Azab
- a Department of Pharmaceutical Chemistry , College of Pharmacy, King Saud University , Riyadh , Saudi Arabia .,e Department of Organic Chemistry , Faculty of Pharmacy, Al-Azhar University , Cairo , Egypt
| |
Collapse
|
29
|
Lambrinidis G, Vallianatou T, Tsantili-Kakoulidou A. In vitro, in silico and integrated strategies for the estimation of plasma protein binding. A review. Adv Drug Deliv Rev 2015; 86:27-45. [PMID: 25819487 DOI: 10.1016/j.addr.2015.03.011] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 02/11/2015] [Accepted: 03/20/2015] [Indexed: 12/28/2022]
Abstract
Plasma protein binding (PPB) strongly affects drug distribution and pharmacokinetic behavior with consequences in overall pharmacological action. Extended plasma protein binding may be associated with drug safety issues and several adverse effects, like low clearance, low brain penetration, drug-drug interactions, loss of efficacy, while influencing the fate of enantiomers and diastereoisomers by stereoselective binding within the body. Therefore in holistic drug design approaches, where ADME(T) properties are considered in parallel with target affinity, considerable efforts are focused in early estimation of PPB mainly in regard to human serum albumin (HSA), which is the most abundant and most important plasma protein. The second critical serum protein α1-acid glycoprotein (AGP), although often underscored, plays also an important and complicated role in clinical therapy and thus the last years it has been studied thoroughly too. In the present review, after an overview of the principles of HSA and AGP binding as well as the structure topology of the proteins, the current trends and perspectives in the field of PPB predictions are presented and discussed considering both HSA and AGP binding. Since however for the latter protein systematic studies have started only the last years, the review focuses mainly to HSA. One part of the review highlights the challenge to develop rapid techniques for HSA and AGP binding simulation and their performance in assessment of PPB. The second part focuses on in silico approaches to predict HSA and AGP binding, analyzing and evaluating structure-based and ligand-based methods, as well as combination of both methods in the aim to exploit the different information and overcome the limitations of each individual approach. Ligand-based methods use the Quantitative Structure-Activity Relationships (QSAR) methodology to establish quantitate models for the prediction of binding constants from molecular descriptors, while they provide only indirect information on binding mechanism. Efforts for the establishment of global models, automated workflows and web-based platforms for PPB predictions are presented and discussed. Structure-based methods relying on the crystal structures of drug-protein complexes provide detailed information on the underlying mechanism but are usually restricted to specific compounds. They are useful to identify the specific binding site while they may be important in investigating drug-drug interactions, related to PPB. Moreover, chemometrics or structure-based modeling may be supported by experimental data a promising integrated alternative strategy for ADME(T) properties optimization. In the case of PPB the use of molecular modeling combined with bioanalytical techniques is frequently used for the investigation of AGP binding.
Collapse
|
30
|
Pires DEV, Blundell TL, Ascher DB. pkCSM: Predicting Small-Molecule Pharmacokinetic and Toxicity Properties Using Graph-Based Signatures. J Med Chem 2015; 58:4066-72. [PMID: 25860834 PMCID: PMC4434528 DOI: 10.1021/acs.jmedchem.5b00104] [Citation(s) in RCA: 2455] [Impact Index Per Article: 245.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
![]()
Drug development has a high attrition
rate, with poor pharmacokinetic
and safety properties a significant hurdle. Computational approaches
may help minimize these risks. We have developed a novel approach
(pkCSM) which uses graph-based signatures to develop predictive models
of central ADMET properties for drug development. pkCSM performs as
well or better than current methods. A freely accessible web server
(http://structure.bioc.cam.ac.uk/pkcsm), which retains
no information submitted to it, provides an integrated platform to
rapidly evaluate pharmacokinetic and toxicity properties.
Collapse
Affiliation(s)
- Douglas E V Pires
- †Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Sanger Building, Cambridge, Cambridgshire CB2 1GA, U.K.,‡Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-002, Brazil
| | - Tom L Blundell
- †Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Sanger Building, Cambridge, Cambridgshire CB2 1GA, U.K
| | - David B Ascher
- †Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Sanger Building, Cambridge, Cambridgshire CB2 1GA, U.K
| |
Collapse
|
31
|
Maltarollo VG, Gertrudes JC, Oliveira PR, Honorio KM. Applying machine learning techniques for ADME-Tox prediction: a review. Expert Opin Drug Metab Toxicol 2014; 11:259-71. [PMID: 25440524 DOI: 10.1517/17425255.2015.980814] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Pharmacokinetics involves the study of absorption, distribution, metabolism, excretion and toxicity of xenobiotics (ADME-Tox). In this sense, the ADME-Tox profile of a bioactive compound can impact its efficacy and safety. Moreover, efficacy and safety were considered some of the major causes of clinical failures in the development of new chemical entities. In this context, machine learning (ML) techniques have been often used in ADME-Tox studies due to the existence of compounds with known pharmacokinetic properties available for generating predictive models. AREAS COVERED This review examines the growth in the use of some ML techniques in ADME-Tox studies, in particular supervised and unsupervised techniques. Also, some critical points (e.g., size of the data set and type of output variable) must be considered during the generation of models that relate ADME-Tox properties and biological activity. EXPERT OPINION ML techniques have been successfully employed in pharmacokinetic studies, helping the complex process of designing new drug candidates from the use of reliable ML models. An application of this procedure would be the prediction of ADME-Tox properties from studies of quantitative structure-activity relationships or the discovery of new compounds from a virtual screening using filters based on results obtained from ML techniques.
Collapse
Affiliation(s)
- Vinícius Gonçalves Maltarollo
- Federal University of ABC (UFABC), Centre for Natural Sciences and Humanities , Santa Adélia Street, 166, Bangu, Santo André -SP , Brazil
| | | | | | | |
Collapse
|
32
|
|
33
|
Fotaki N. Pros and cons of methods used for the prediction of oral drug absorption. Expert Rev Clin Pharmacol 2014; 2:195-208. [DOI: 10.1586/17512433.2.2.195] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
34
|
de Toledo JS, Junior PES, Manfrim V, Pinzan CF, de Araujo AS, Cruz AK, Emery FS. Synthesis, cytotoxicity and in vitro antileishmanial activity of naphthothiazoles. Chem Biol Drug Des 2014; 81:749-56. [PMID: 23421616 DOI: 10.1111/cbdd.12123] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 12/11/2012] [Accepted: 02/12/2013] [Indexed: 01/23/2023]
Abstract
The leishmaniasis is a spectral disease caused by the protozoan Leishmania spp., which threatens millions of people worldwide. Current treatments exhibit high toxicity, and there is no vaccine available. The need for new lead compounds with leishmanicidal activity is urgent. Considering that many lead leishmanicidal compounds contain a quinoidal scaffold and the thiazole heterocyclic ring is found in a number of antimicrobial drugs, we proposed a hybridization approach to generate a diverse set of semi-synthetic heterocycles with antileishmanial activity. We found that almost all synthesized compounds demonstrated potent activity against promastigotes of Leishmania (Viannia) braziliensis and reduced the survival index of Leishmania amastigotes in mammalian macrophages. Furthermore, the compounds were not cytotoxic to macrophages at fivefold higher concentrations than the EC50 for promastigotes. All molecules fulfilled Lipinski's Rule of Five, which predicts efficient orally absorption and permeation through biological membranes, the in silico pharmacokinetic profile confirmed these characteristics. The potent and selective activity of semi-synthetic naphthothiazoles against promastigotes and amastigotes reveals that the 2-amino-naphthothiazole ring may represent a scaffold for the design of compounds with leishmanicidal properties and encourage the development of drug formulation and new compounds for further studies in vivo.
Collapse
Affiliation(s)
- Juliano S de Toledo
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP 14049-900, Brazil
| | | | | | | | | | | | | |
Collapse
|
35
|
Fang J, Yang R, Gao L, Zhou D, Yang S, Liu AL, Du GH. Predictions of BuChE inhibitors using support vector machine and naive Bayesian classification techniques in drug discovery. J Chem Inf Model 2013; 53:3009-20. [PMID: 24144102 DOI: 10.1021/ci400331p] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Butyrylcholinesterase (BuChE, EC 3.1.1.8) is an important pharmacological target for Alzheimer's disease (AD) treatment. However, the currently available BuChE inhibitor screening assays are expensive, labor-intensive, and compound-dependent. It is necessary to develop robust in silico methods to predict the activities of BuChE inhibitors for the lead identification. In this investigation, support vector machine (SVM) models and naive Bayesian models were built to discriminate BuChE inhibitors (BuChEIs) from the noninhibitors. Each molecule was initially represented in 1870 structural descriptors (1235 from ADRIANA.Code, 334 from MOE, and 301 from Discovery studio). Correlation analysis and stepwise variable selection method were applied to figure out activity-related descriptors for prediction models. Additionally, structural fingerprint descriptors were added to improve the predictive ability of models, which were measured by cross-validation, a test set validation with 1001 compounds and an external test set validation with 317 diverse chemicals. The best two models gave Matthews correlation coefficient of 0.9551 and 0.9550 for the test set and 0.9132 and 0.9221 for the external test set. To demonstrate the practical applicability of the models in virtual screening, we screened an in-house data set with 3601 compounds, and 30 compounds were selected for further bioactivity assay. The assay results showed that 10 out of 30 compounds exerted significant BuChE inhibitory activities with IC50 values ranging from 0.32 to 22.22 μM, at which three new scaffolds as BuChE inhibitors were identified for the first time. To our best knowledge, this is the first report on BuChE inhibitors using machine learning approaches. The models generated from SVM and naive Bayesian approaches successfully predicted BuChE inhibitors. The study proved the feasibility of a new method for predicting bioactivities of ligands and discovering novel lead compounds.
Collapse
Affiliation(s)
- Jiansong Fang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing 100050, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
36
|
Law V, Knox C, Djoumbou Y, Jewison T, Guo AC, Liu Y, Maciejewski A, Arndt D, Wilson M, Neveu V, Tang A, Gabriel G, Ly C, Adamjee S, Dame ZT, Han B, Zhou Y, Wishart DS. DrugBank 4.0: shedding new light on drug metabolism. Nucleic Acids Res 2013; 42:D1091-7. [PMID: 24203711 PMCID: PMC3965102 DOI: 10.1093/nar/gkt1068] [Citation(s) in RCA: 1465] [Impact Index Per Article: 122.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
DrugBank (http://www.drugbank.ca) is a comprehensive online database containing extensive biochemical and pharmacological information about drugs, their mechanisms and their targets. Since it was first described in 2006, DrugBank has rapidly evolved, both in response to user requests and in response to changing trends in drug research and development. Previous versions of DrugBank have been widely used to facilitate drug and in silico drug target discovery. The latest update, DrugBank 4.0, has been further expanded to contain data on drug metabolism, absorption, distribution, metabolism, excretion and toxicity (ADMET) and other kinds of quantitative structure activity relationships (QSAR) information. These enhancements are intended to facilitate research in xenobiotic metabolism (both prediction and characterization), pharmacokinetics, pharmacodynamics and drug design/discovery. For this release, >1200 drug metabolites (including their structures, names, activity, abundance and other detailed data) have been added along with >1300 drug metabolism reactions (including metabolizing enzymes and reaction types) and dozens of drug metabolism pathways. Another 30 predicted or measured ADMET parameters have been added to each DrugCard, bringing the average number of quantitative ADMET values for Food and Drug Administration-approved drugs close to 40. Referential nuclear magnetic resonance and MS spectra have been added for almost 400 drugs as well as spectral and mass matching tools to facilitate compound identification. This expanded collection of drug information is complemented by a number of new or improved search tools, including one that provides a simple analyses of drug–target, –enzyme and –transporter associations to provide insight on drug–drug interactions.
Collapse
Affiliation(s)
- Vivian Law
- Department of Computing Science, University of Alberta, Edmonton, AB, Canada T6G 2E8, Department Biological Sciences, University of Alberta, Edmonton, AB, Canada T6G 2E8, Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada T6G 2N8 and National Institute for Nanotechnology, 11421 Saskatchewan Drive, Edmonton, AB, Canada T6G 2M9
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Inhibitors of Trypanosoma brucei trypanothione reductase: comparative molecular field analysis modeling and structural basis for selective inhibition. Future Med Chem 2013; 5:1753-62. [DOI: 10.4155/fmc.13.140] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background: Sleeping sickness is a major cause of death in Africa. Since no secure treatment is available, the development of novel therapeutic agents is urgent. In this context, the enzyme trypanothione reductase (TR) is a prominent molecular target that has been investigated in drug design for sleeping sickness. Results: In this study, comparative molecular field analysis models were generated for a series of Trypanosoma brucei TR inhibitors. Statistically significant results were obtained and the models were applied to predict the activity of external test sets, with good correlation between predicted and experimental results. We have also investigated the structural requirements for the selective inhibition of the parasite‘s enzyme over the human glutathione reductase. Conclusion: The quantitative structure–activity relationship models provided valuable information regarding the essential molecular requirements for the inhibitory activity upon the target protein, providing important insights into the design of more potent and selective TR inhibitors.
Collapse
|
38
|
da Mota EG, Silva DG, Guimarães MC, da Cunha EF, Freitas MP. Computer-assisted design of novel 1,4-dihydropyridine calcium channel blockers. MOLECULAR SIMULATION 2013. [DOI: 10.1080/08927022.2013.829220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
39
|
|
40
|
Zhu XW, Sedykh A, Zhu H, Liu SS, Tropsha A. The use of pseudo-equilibrium constant affords improved QSAR models of human plasma protein binding. Pharm Res 2013; 30:1790-8. [PMID: 23568522 DOI: 10.1007/s11095-013-1023-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 03/04/2013] [Indexed: 12/21/2022]
Abstract
PURPOSE To develop accurate in silico predictors of Plasma Protein Binding (PPB). METHODS Experimental PPB data were compiled for over 1,200 compounds. Two endpoints have been considered: (1) fraction bound (%PPB); and (2) the logarithm of a pseudo binding constant (lnKa) derived from %PPB. The latter metric was employed because it reflects the PPB thermodynamics and the distribution of the transformed data is closer to normal. Quantitative Structure-Activity Relationship (QSAR) models were built with Dragon descriptors and three statistical methods. RESULTS Five-fold external validation procedure resulted in models with the prediction accuracy (R²) of 0.67 ± 0.04 and 0.66 ± 0.04, respectively, and the mean absolute error (MAE) of 15.3 ± 0.2% and 13.6 ± 0.2%, respectively. Models were validated with two external datasets: 173 compounds from DrugBank, and 236 chemicals from the US EPA ToxCast project. Models built with lnKa were significantly more accurate (MAE of 6.2-10.7 %) than those built with %PPB (MAE of 11.9-17.6 %) for highly bound compounds both for the training and the external sets. CONCLUSIONS The pseudo binding constant (lnKa) is more appropriate for characterizing PPB binding than conventional %PPB. Validated QSAR models developed herein can be applied as reliable tools in early drug development and in chemical risk assessment.
Collapse
Affiliation(s)
- Xiang-Wei Zhu
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science & Engineering, Tongji University, 417 Mingjing Building, Shanghai 200092, China
| | | | | | | | | |
Collapse
|
41
|
Valli M, dos Santos RN, Figueira LD, Nakajima CH, Castro-Gamboa I, Andricopulo AD, Bolzani VS. Development of a natural products database from the biodiversity of Brazil. JOURNAL OF NATURAL PRODUCTS 2013; 76:439-44. [PMID: 23330984 DOI: 10.1021/np3006875] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
We describe herein the design and development of an innovative tool called the NuBBE database (NuBBEDB), a new Web-based database, which incorporates several classes of secondary metabolites and derivatives from the biodiversity of Brazil. This natural product database incorporates botanical, chemical, pharmacological, and toxicological compound information. The NuBBEDB provides specialized information to the worldwide scientific community and can serve as a useful tool for studies on the multidisciplinary interfaces related to chemistry and biology, including virtual screening, dereplication, metabolomics, and medicinal chemistry. The NuBBEDB site is at http://nubbe.iq.unesp.br/nubbeDB.html .
Collapse
Affiliation(s)
- Marilia Valli
- Núcleo de Bioensaios, Biossíntese e Ecofisiologia de Produtos Naturais (NuBBE), Departamento de Química Orgânica, Instituto de Química, UNESP - Univ. Estadual Paulista, 14801-970, Araraquara-SP, Brazil
| | | | | | | | | | | | | |
Collapse
|
42
|
Cheng F, Li W, Zhou Y, Shen J, Wu Z, Liu G, Lee PW, Tang Y. admetSAR: a comprehensive source and free tool for assessment of chemical ADMET properties. J Chem Inf Model 2012; 52:3099-105. [PMID: 23092397 DOI: 10.1021/ci300367a] [Citation(s) in RCA: 1214] [Impact Index Per Article: 93.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties play key roles in the discovery/development of drugs, pesticides, food additives, consumer products, and industrial chemicals. This information is especially useful when to conduct environmental and human hazard assessment. The most critical rate limiting step in the chemical safety assessment workflow is the availability of high quality data. This paper describes an ADMET structure-activity relationship database, abbreviated as admetSAR. It is an open source, text and structure searchable, and continually updated database that collects, curates, and manages available ADMET-associated properties data from the published literature. In admetSAR, over 210,000 ADMET annotated data points for more than 96,000 unique compounds with 45 kinds of ADMET-associated properties, proteins, species, or organisms have been carefully curated from a large number of diverse literatures. The database provides a user-friendly interface to query a specific chemical profile, using either CAS registry number, common name, or structure similarity. In addition, the database includes 22 qualitative classification and 5 quantitative regression models with highly predictive accuracy, allowing to estimate ecological/mammalian ADMET properties for novel chemicals. AdmetSAR is accessible free of charge at http://www.admetexp.org.
Collapse
Affiliation(s)
- Feixiong Cheng
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Cao D, Wang J, Zhou R, Li Y, Yu H, Hou T. ADMET evaluation in drug discovery. 11. PharmacoKinetics Knowledge Base (PKKB): a comprehensive database of pharmacokinetic and toxic properties for drugs. J Chem Inf Model 2012; 52:1132-7. [PMID: 22559792 DOI: 10.1021/ci300112j] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Good and extensive experimental ADMET (absorption, distribution, metabolism, excretion, and toxicity) data is critical for developing reliable in silico ADMET models. Here we develop a PharmacoKinetics Knowledge Base (PKKB) to compile comprehensive information about ADMET properties into a single electronic repository. We incorporate more than 10 000 experimental ADMET measurements of 1685 drugs into the PKKB. The ADMET properties in the PKKB include octanol/water partition coefficient, solubility, dissociation constant, intestinal absorption, Caco-2 permeability, human bioavailability, plasma protein binding, blood-plasma partitioning ratio, volume of distribution, metabolism, half-life, excretion, urinary excretion, clearance, toxicity, half lethal dose in rat or mouse, etc. The PKKB provides the most extensive collection of freely available data for ADMET properties up to date. All these ADMET properties, as well as the pharmacological information and the calculated physiochemical properties are integrated into a web-based information system. Eleven separated data sets for octanol/water partition coefficient, solubility, blood-brain partitioning, intestinal absorption, Caco-2 permeability, human oral bioavailability, and P-glycoprotein inhibitors have been provided for free download and can be used directly for ADMET modeling. The PKKB is available online at http://cadd.suda.edu.cn/admet.
Collapse
Affiliation(s)
- Dongyue Cao
- Institute of Functional Nano & Soft Materials-FUNSOM and Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | | | | | | | | | | |
Collapse
|
44
|
Moda TL, Andricopulo AD. Consensus hologram QSAR modeling for the prediction of human intestinal absorption. Bioorg Med Chem Lett 2012; 22:2889-93. [DOI: 10.1016/j.bmcl.2012.02.061] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 02/16/2012] [Accepted: 02/17/2012] [Indexed: 11/28/2022]
|
45
|
Silva DG, Freitas MP, da Cunha EFF, Ramalho TC, Nunes CA. Rational design of small modified peptides as ACE inhibitors. MEDCHEMCOMM 2012. [DOI: 10.1039/c2md20214j] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
46
|
Singh S, Gupta SK, Nischal A, Khattri S, Nath R, Pant KK, Seth PK. Identification and Characterization of Novel Small-Molecule Inhibitors against Hepatitis Delta Virus Replication by Using Docking Strategies. HEPATITIS MONTHLY 2011; 11:803-809. [DOI: 10.5812/kowsar.1735143x.1387] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
|
47
|
Andrade CH, Freitas LMD, Oliveira VD. Twenty-six years of HIV science: an overview of anti-HIV drugs metabolism. BRAZ J PHARM SCI 2011. [DOI: 10.1590/s1984-82502011000200003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
From the identification of HIV as the agent causing AIDS, to the development of effective antiretroviral drugs, the scientific achievements in HIV research over the past twenty-six years have been formidable. Currently, there are twenty-five anti-HIV compounds which have been formally approved for clinical use in the treatment of AIDS. These compounds fall into six categories: nucleoside reverse transcriptase inhibitors (NRTIs), nucleotide reverse transcriptase inhibitors (NtRTIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs), protease inhibitors (PIs), cell entry inhibitors or fusion inhibitors (FIs), co-receptor inhibitors (CRIs), and integrase inhibitors (INIs). Metabolism by the host organism is one of the most important determinants of the pharmacokinetic profile of a drug. Formation of active or toxic metabolites will also have an impact on the pharmacological and toxicological outcomes. Therefore, it is widely recognized that metabolism studies of a new chemical entity need to be addressed early in the drug discovery process. This paper describes an overview of the metabolism of currently available anti-HIV drugs.
Collapse
|
48
|
El-Deeb IM, Bayoumi SM, El-Sherbeny MA, Abdel-Aziz AAM. Synthesis and antitumor evaluation of novel cyclic arylsulfonylureas: ADME-T and pharmacophore prediction. Eur J Med Chem 2010; 45:2516-30. [PMID: 20236733 DOI: 10.1016/j.ejmech.2010.02.038] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2009] [Revised: 02/11/2010] [Accepted: 02/15/2010] [Indexed: 11/25/2022]
|
49
|
Salum LB, Andricopulo AD. Fragment-based QSAR strategies in drug design. Expert Opin Drug Discov 2010; 5:405-12. [DOI: 10.1517/17460441003782277] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
50
|
GRABOWSKI T, JAROSZEWSKI JJ. Bioavailability of veterinary drugsin vivoandin silico. J Vet Pharmacol Ther 2009; 32:249-57. [DOI: 10.1111/j.1365-2885.2008.01034.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|