1
|
Buljan A, Stepanić V, Čikoš A, Babić Brčić S, Bojanić K, Roje M. Total Synthesis and Biological Profiling of Putative (±)-Marinoaziridine B and (±)- N-Methyl Marinoaziridine A. Mar Drugs 2024; 22:310. [PMID: 39057419 PMCID: PMC11278217 DOI: 10.3390/md22070310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
The total synthesis of two new marine natural products, (±)-marinoaziridine B 7 and (±)-N-methyl marinoaziridine A 8, was accomplished. The (±)-marinoaziridine 7 was prepared in a six-step linear sequence with a 2% overall yield. The key steps in our strategy were the preparation of the chiral epoxide (±)-5 using the Johnson Corey Chaykovsky reaction, followed by the ring-opening reaction and the Staudinger reaction. The N,N-dimethylation of compound (±)-7 gives (±)-N-methyl marinoaziridine A 8. The NMR spectra of synthetized (±)-marinoaziridine B 7 and isolated natural product did not match. The compounds are biologically characterized using relevant in silico, in vitro and in vivo methods. In silico ADMET and bioactivity profiling predicted toxic and neuromodulatory effects. In vitro screening by MTT assay on three cell lines (MCF-7, H-460, HEK293T) showed that both compounds exhibited moderate to strong antiproliferative and cytotoxic effects. Antimicrobial tests on bacterial cultures of Escherichia coli and Staphylococcus aureus demonstrated the dose-dependent inhibition of the growth of both bacteria. In vivo toxicological tests were performed on zebrafish Danio rerio and showed a significant reduction of zebrafish mortality due to N-methylation in (±)-8.
Collapse
Affiliation(s)
- Anđela Buljan
- Laboratory for Chiral Technologies, Scientific Center of Excellence for Marine Bioprospecting-BioProCro, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia;
| | - Višnja Stepanić
- Laboratory for Machine Learning and Knowledge Representation, Division of Electronics, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia;
| | - Ana Čikoš
- NMR Centre, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia;
| | - Sanja Babić Brčić
- Laboratory for Aquaculture Biotechnology, Scientific Center of Excellence for Marine Bioprospecting-BioProCro, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia; (S.B.B.); (K.B.)
| | - Krunoslav Bojanić
- Laboratory for Aquaculture Biotechnology, Scientific Center of Excellence for Marine Bioprospecting-BioProCro, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia; (S.B.B.); (K.B.)
| | - Marin Roje
- Laboratory for Chiral Technologies, Scientific Center of Excellence for Marine Bioprospecting-BioProCro, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia;
| |
Collapse
|
2
|
Schimunek J, Seidl P, Elez K, Hempel T, Le T, Noé F, Olsson S, Raich L, Winter R, Gokcan H, Gusev F, Gutkin EM, Isayev O, Kurnikova MG, Narangoda CH, Zubatyuk R, Bosko IP, Furs KV, Karpenko AD, Kornoushenko YV, Shuldau M, Yushkevich A, Benabderrahmane MB, Bousquet-Melou P, Bureau R, Charton B, Cirou BC, Gil G, Allen WJ, Sirimulla S, Watowich S, Antonopoulos N, Epitropakis N, Krasoulis A, Itsikalis V, Theodorakis S, Kozlovskii I, Maliutin A, Medvedev A, Popov P, Zaretckii M, Eghbal-Zadeh H, Halmich C, Hochreiter S, Mayr A, Ruch P, Widrich M, Berenger F, Kumar A, Yamanishi Y, Zhang KYJ, Bengio E, Bengio Y, Jain MJ, Korablyov M, Liu CH, Marcou G, Glaab E, Barnsley K, Iyengar SM, Ondrechen MJ, Haupt VJ, Kaiser F, Schroeder M, Pugliese L, Albani S, Athanasiou C, Beccari A, Carloni P, D'Arrigo G, Gianquinto E, Goßen J, Hanke A, Joseph BP, Kokh DB, Kovachka S, Manelfi C, Mukherjee G, Muñiz-Chicharro A, Musiani F, Nunes-Alves A, Paiardi G, Rossetti G, Sadiq SK, Spyrakis F, Talarico C, Tsengenes A, Wade RC, Copeland C, Gaiser J, Olson DR, Roy A, Venkatraman V, Wheeler TJ, Arthanari H, Blaschitz K, Cespugli M, Durmaz V, Fackeldey K, Fischer PD, Gorgulla C, Gruber C, Gruber K, Hetmann M, Kinney JE, Padmanabha Das KM, Pandita S, Singh A, Steinkellner G, Tesseyre G, Wagner G, Wang ZF, Yust RJ, Druzhilovskiy DS, Filimonov DA, Pogodin PV, Poroikov V, Rudik AV, Stolbov LA, Veselovsky AV, De Rosa M, De Simone G, Gulotta MR, Lombino J, Mekni N, Perricone U, Casini A, Embree A, Gordon DB, Lei D, Pratt K, Voigt CA, Chen KY, Jacob Y, Krischuns T, Lafaye P, Zettor A, Rodríguez ML, White KM, Fearon D, Von Delft F, Walsh MA, Horvath D, Brooks CL, Falsafi B, Ford B, García-Sastre A, Yup Lee S, Naffakh N, Varnek A, Klambauer G, Hermans TM. A community effort in SARS-CoV-2 drug discovery. Mol Inform 2024; 43:e202300262. [PMID: 37833243 PMCID: PMC11299051 DOI: 10.1002/minf.202300262] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 10/15/2023]
Abstract
The COVID-19 pandemic continues to pose a substantial threat to human lives and is likely to do so for years to come. Despite the availability of vaccines, searching for efficient small-molecule drugs that are widely available, including in low- and middle-income countries, is an ongoing challenge. In this work, we report the results of an open science community effort, the "Billion molecules against COVID-19 challenge", to identify small-molecule inhibitors against SARS-CoV-2 or relevant human receptors. Participating teams used a wide variety of computational methods to screen a minimum of 1 billion virtual molecules against 6 protein targets. Overall, 31 teams participated, and they suggested a total of 639,024 molecules, which were subsequently ranked to find 'consensus compounds'. The organizing team coordinated with various contract research organizations (CROs) and collaborating institutions to synthesize and test 878 compounds for biological activity against proteases (Nsp5, Nsp3, TMPRSS2), nucleocapsid N, RdRP (only the Nsp12 domain), and (alpha) spike protein S. Overall, 27 compounds with weak inhibition/binding were experimentally identified by binding-, cleavage-, and/or viral suppression assays and are presented here. Open science approaches such as the one presented here contribute to the knowledge base of future drug discovery efforts in finding better SARS-CoV-2 treatments.
Collapse
|
3
|
Tallei TE, Fatimawali, Adam AA, Ekatanti D, Celik I, Fatriani R, Nainu F, Kusuma WA, Rabaan AA, Idroes R. Molecular insights into the anti-inflammatory activity of fermented pineapple juice using multimodal computational studies. Arch Pharm (Weinheim) 2024; 357:e2300422. [PMID: 37861276 DOI: 10.1002/ardp.202300422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/29/2023] [Accepted: 10/04/2023] [Indexed: 10/21/2023]
Abstract
Pineapple has been recognized for its potential to enhance health and well-being. This study aimed to gain molecular insights into the anti-inflammatory properties of fermented pineapple juice using multimodal computational studies. In this study, pineapple juice was fermented using Lactobacillus paracasei, and the solution underwent liquid chromatography-mass spectrometry analysis. Network pharmacology was applied to investigate compound interactions and targets. In silico methods assessed compound bioactivities. Protein-protein interactions, network topology, and enrichment analysis identified key compounds. Molecular docking explored compound-receptor interactions in inflammation regulation. Molecular dynamics simulations were conducted to confirm the stability of interactions between the identified crucial compounds and their respective receptors. The study revealed several compounds including short-chain fatty acids, peptides, dihydroxyeicosatrienoic acids, and glycerides that exhibited promising anti-inflammatory properties. Leucyl-leucyl-norleucine and Leu-Leu-Tyr exhibited robust and stable interactions with mitogen-activated protein kinase 14 and IκB kinase β, respectively, indicating their potential as promising therapeutic agents for inflammation modulation. This proposition is grounded in the pivotal involvement of these two proteins in inflammatory signaling pathways. These findings provide valuable insights into the anti-inflammatory potential of these compounds, serving as a foundation for further experimental validation and exploration. Future studies can build upon these results to advance the development of these compounds as effective anti-inflammatory agents.
Collapse
Grants
- 053/E5/PG.02.00.PL/2023 Directorate of Research, Technology, and Community Service of the Ministry of Education, Culture, Research, and Technology, Republic of Indonesia
- 189/UN12.13/LT/2023 Directorate of Research, Technology, and Community Service of the Ministry of Education, Culture, Research, and Technology, Republic of Indonesia
Collapse
Affiliation(s)
- Trina Ekawati Tallei
- Department of Biology, Faculty of Mathematics and Natural Science, Sam Ratulangi University, Manado, North Sulawesi, Indonesia
| | - Fatimawali
- Pharmacy Study Program, Faculty of Mathematics and Natural Science, Sam Ratulangi University, Manado, North Sulawesi, Indonesia
| | - Ahmad Akroman Adam
- Dentistry Study Program, Faculty of Medicine, Sam Ratulangi University, Manado, North Sulawesi, Indonesia
| | - Dewi Ekatanti
- Pharmacy Study Program, Faculty of Mathematics and Natural Science, Sam Ratulangi University, Manado, North Sulawesi, Indonesia
| | - Ismail Celik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey
| | - Rizka Fatriani
- Tropical Biopharmaca Research Center, IPB University, Bogor, Indonesia
| | - Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Wisnu Ananta Kusuma
- Tropical Biopharmaca Research Center, IPB University, Bogor, Indonesia
- Department of Computer Science, Faculty of Mathematics and Natural Sciences, IPB University, Bogor, Indonesia
| | - Ali A Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur, Pakistan
| | - Rinaldi Idroes
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, Universitas Syiah Kuala, Kopelma Darussalam, Banda Aceh, Aceh, Indonesia
| |
Collapse
|
4
|
Bender AM, Parr LC, Livingston WB, Lindsley CW, Merryman WD. 2B Determined: The Future of the Serotonin Receptor 2B in Drug Discovery. J Med Chem 2023; 66:11027-11039. [PMID: 37584406 PMCID: PMC11073569 DOI: 10.1021/acs.jmedchem.3c01178] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
The cardiotoxicity associated with des-ethyl-dexfenfluramine (norDF) and related agonists of the serotonin receptor 2B (5-HT2B) has solidified the receptor's place as an "antitarget" in drug discovery. Conversely, a growing body of evidence has highlighted the utility of 5-HT2B antagonists for the treatment of pulmonary arterial hypertension (PAH), valvular heart disease (VHD), and related cardiopathies. In this Perspective, we summarize the link between the clinical failure of fenfluramine-phentermine (fen-phen) and the subsequent research on the role of 5-HT2B in disease progression, as well as the development of drug-like and receptor subtype-selective 5-HT2B antagonists. Such agents represent a promising class for the treatment of PAH and VHD, but their utility has been historically understudied due to the clinical disasters associated with 5-HT2B. Herein, it is our aim to examine the current state of 5-HT2B drug discovery, with an emphasis on the receptor's role in the central nervous system (CNS) versus the periphery.
Collapse
Affiliation(s)
- Aaron M Bender
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Lauren C Parr
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - William B Livingston
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37240, United States
| | - Craig W Lindsley
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37240, United States
| |
Collapse
|
5
|
Ferreira MKA, Freitas WPO, Barbosa IM, da Rocha MN, da Silva AW, de Lima Rebouças E, da Silva Mendes FR, Alves CR, Nunes PIG, Marinho MM, Furtado RF, Santos FA, Marinho ES, de Menezes JESA, dos Santos HS. Heterocyclic chalcone ( E)-1-(2-hydroxy-3,4,6-trimethoxyphenyl)-3-(thiophen-2-yl) prop-2-en-1-one derived from a natural product with antinociceptive, anti-inflammatory, and hypoglycemic effect in adult zebrafish. 3 Biotech 2023; 13:276. [PMID: 37457871 PMCID: PMC10349009 DOI: 10.1007/s13205-023-03696-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023] Open
Abstract
Diabetes is a disease linked to pathologies, such as chronic inflammation, neuropathy, and pain. The synthesis by the Claisen-Schmidt condensation reaction aims to obtain medium to high yield chalconic derivatives. Studies for the synthesis of new chalcone molecules aim at the structural manipulation of aromatic rings, as well as the replacement of rings by heterocycles, and combination through chemical reactions of synthesized structures with other molecules, in order to enhance biological activity. A chalcone was synthesized and evaluated for its antinociceptive, anti-inflammatory and hypoglycemic effect in adult zebrafish. In addition to reducing nociceptive behavior, chalcone (40 mg/kg) reversed post-treatment-induced acute and chronic hyperglycemia and reduced carrageenan-induced abdominal edema in zebrafish. It also showed an inhibitory effect on NO production in J774A.1 cells. When compared with the control groups, the oxidative stress generated after chronic hyperglycemia and after induction of abdominal edema was significantly reduced by chalcone. Molecular docking simulations of chalcone with Cox -1, Cox-2, and TRPA1 channel enzymes were performed and indicated that chalcone has a higher affinity for the COX-1 enzyme and 4 interactions with the TRPA1 channel. Chalcone also showed good pharmacokinetic properties as assessed by ADMET. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-023-03696-8.
Collapse
Affiliation(s)
- Maria Kueirislene Amancio Ferreira
- Laboratório de Química de Produtos Naturais-LQPNS, Universidade Estadual do Ceará, Programa de Pós-Graduação em Ciências Naturais, Fortaleza, CE Brazil
| | - Wendy Pascoal Oliveira Freitas
- Laboratório de Química de Produtos Naturais-LQPNS, Universidade Estadual do Ceará, Programa de Pós-Graduação em Ciências Naturais, Fortaleza, CE Brazil
| | - Italo Moura Barbosa
- Laboratório de Química de Produtos Naturais-LQPNS, Universidade Estadual do Ceará, Programa de Pós-Graduação em Ciências Naturais, Fortaleza, CE Brazil
| | - Matheus Nunes da Rocha
- Laboratório de Química de Produtos Naturais-LQPNS, Universidade Estadual do Ceará, Programa de Pós-Graduação em Ciências Naturais, Fortaleza, CE Brazil
| | - Antônio Wlisses da Silva
- Programa de Doutorado em Biotecnologia, Rede Nordeste de Biotecnologia (RENORBIO), Fortaleza, CE Brazil
| | - Emanuela de Lima Rebouças
- Programa de Doutorado em Biotecnologia, Rede Nordeste de Biotecnologia (RENORBIO), Fortaleza, CE Brazil
| | | | - Carlucio Roberto Alves
- Laboratório de Sistemas de Nanotecnologia e BiomateriaisPrograma de Pós-Graduação em Ciências Naturais, Universidade Estadual do Ceará, Fortaleza, CE Brazil
| | - Paulo Iury Gomes Nunes
- Departamento de Fisiologia e Farmacologia Laboratório de Produtos Naturais, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE Brazil
| | | | | | - Flávia Almeida Santos
- Departamento de Fisiologia e Farmacologia Laboratório de Produtos Naturais, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE Brazil
| | - Emmanuel Silva Marinho
- Laboratório de Química de Produtos Naturais-LQPNS, Universidade Estadual do Ceará, Programa de Pós-Graduação em Ciências Naturais, Fortaleza, CE Brazil
| | - Jane Eire Silva Alencar de Menezes
- Laboratório de Química de Produtos Naturais-LQPNS, Universidade Estadual do Ceará, Programa de Pós-Graduação em Ciências Naturais, Fortaleza, CE Brazil
| | - Helcio Silva dos Santos
- Laboratório de Química de Produtos Naturais-LQPNS, Universidade Estadual do Ceará, Programa de Pós-Graduação em Ciências Naturais, Fortaleza, CE Brazil
- Programa de Doutorado em Biotecnologia, Rede Nordeste de Biotecnologia (RENORBIO), Fortaleza, CE Brazil
- Departamento de Química, Universidade Estadual Vale do Acaraú, Sobral, CE Brazil
| |
Collapse
|
6
|
Khaled DM, Elshakre ME, Noamaan MA, Butt H, Abdel Fattah MM, Gaber DA. A Computational QSAR, Molecular Docking and In Vitro Cytotoxicity Study of Novel Thiouracil-Based Drugs with Anticancer Activity against Human-DNA Topoisomerase II. Int J Mol Sci 2022; 23:11799. [PMID: 36233102 PMCID: PMC9570267 DOI: 10.3390/ijms231911799] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/16/2022] [Accepted: 09/18/2022] [Indexed: 11/30/2022] Open
Abstract
Computational chemistry, molecular docking, and drug design approaches, combined with the biochemical evaluation of the antitumor activity of selected derivatives of the thiouracil-based dihydroindeno pyrido pyrimidines against topoisomerase I and II. The IC50 of other cell lines including the normal human lung cell line W138, lung cancer cell line, A549, breast cancer cell line, MCF-7, cervical cancer, HeLa, and liver cancer cell line HepG2 was evaluated using biochemical methods. The global reactivity descriptors and physicochemical parameters were computed, showing good agreement with the Lipinski and Veber's rules of the drug criteria. The molecular docking study of the ligands with the topoisomerase protein provides the binding sites, binding energies, and deactivation constant for the inhibition pocket. Various biochemical methods were used to evaluate the IC50 of the cell lines. The QSAR model was developed for colorectal cell line HCT as a case study. Four QSAR statistical models were predicted between the IC50 of the colorectal cell line HCT to correlate the anticancer activity and the computed physicochemical and quantum chemical global reactivity descriptors. The predictive power of the models indicates a good correlation between the observed and the predicted activity.
Collapse
Affiliation(s)
- Doaa M. Khaled
- Histology and Cytology Department, Faculty of Medicine, Helwan University, Cairo 11795, Egypt
| | - Mohamed E. Elshakre
- Chemistry Department, Faculty of Science, Cairo University, Cairo 12613, Egypt
| | - Mahmoud A. Noamaan
- Mathematics Department, Faculty of Science, Cairo University, Cairo 12613, Egypt
| | - Haider Butt
- Department of Mechanical Engineering, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Marwa M. Abdel Fattah
- Histology and Cytology Department, Faculty of Medicine, Misr University for Science & Technology, Cairo P.O. Box 77, Egypt
| | - Dalia A. Gaber
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Helwan University, Cairo 11795, Egypt
- Department of Biomedical Sciences, College of Medicine, Gulf Medical University, Ajman 4184, United Arab Emirates
| |
Collapse
|
7
|
Khairullina V, Martynova Y, Safarova I, Sharipova G, Gerchikov A, Limantseva R, Savchenko R. QSPR Modeling and Experimental Determination of the Antioxidant Activity of Some Polycyclic Compounds in the Radical-Chain Oxidation Reaction of Organic Substrates. Molecules 2022; 27:molecules27196511. [PMID: 36235050 PMCID: PMC9572093 DOI: 10.3390/molecules27196511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/28/2022] [Accepted: 09/28/2022] [Indexed: 01/18/2023] Open
Abstract
The present work addresses the quantitative structure−antioxidant activity relationship in a series of 148 sulfur-containing alkylphenols, natural phenols, chromane, betulonic and betulinic acids, and 20-hydroxyecdysone using GUSAR2019 software. Statistically significant valid models were constructed to predict the parameter logk7, where k7 is the rate constant for the oxidation chain termination by the antioxidant molecule. These results can be used to search for new potentially effective antioxidants in virtual libraries and databases and adequately predict logk7 for test samples. A combination of MNA- and QNA-descriptors with three whole molecule descriptors (topological length, topological volume, and lipophilicity) was used to develop six statistically significant valid consensus QSPR models, which have a satisfactory accuracy in predicting logk7 for training and test set structures: R2TR > 0.6; Q2TR > 0.5; R2TS > 0.5. Our theoretical prediction of logk7 for antioxidants AO1 and AO2, based on consensus models agrees well with the experimental value of the measure in this paper. Thus, the descriptor calculation algorithms implemented in the GUSAR2019 software allowed us to model the kinetic parameters of the reactions underlying the liquid-phase oxidation of organic hydrocarbons.
Collapse
Affiliation(s)
- Veronika Khairullina
- Faculty of Chemistry, Bashkir State University, 450076 Ufa, Russia
- Correspondence: ; Tel.: +7-963-906-6567
| | - Yuliya Martynova
- Faculty of Chemistry, Bashkir State University, 450076 Ufa, Russia
| | - Irina Safarova
- Faculty of Chemistry, Bashkir State University, 450076 Ufa, Russia
| | - Gulnaz Sharipova
- Faculty of Chemistry, Bashkir State University, 450076 Ufa, Russia
| | | | - Regina Limantseva
- Institute of Petrochemistry and Catalysis of the Ufa Federal Research Center of the Russian Academy of Sciences, 450075 Ufa, Russia
| | - Rimma Savchenko
- Institute of Petrochemistry and Catalysis of the Ufa Federal Research Center of the Russian Academy of Sciences, 450075 Ufa, Russia
| |
Collapse
|
8
|
Palladium(II) Complexes of Substituted Salicylaldehydes: Synthesis, Characterization and Investigation of Their Biological Profile. Pharmaceuticals (Basel) 2022; 15:ph15070886. [PMID: 35890184 PMCID: PMC9323974 DOI: 10.3390/ph15070886] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/06/2022] [Accepted: 07/14/2022] [Indexed: 02/04/2023] Open
Abstract
Five palladium(II) complexes of substituted salicylaldehydes (X-saloH, X = 4-Et2N (for 1), 3,5-diBr (for 2), 3,5-diCl (for 3), 5-F (for 4) or 4-OMe (for 5)) bearing the general formula [Pd(X-salo)2] were synthesized and structurally characterized. The crystal structure of complex [Pd(4-Et2N-salo)2] was determined by single-crystal X-ray crystallography. The complexes can scavenge 1,1-diphenyl-picrylhydrazyl and 2,2′-azinobis(3-ethylbenzothiazoline-6-sulfonic acid) radicals and reduce H2O2. They are active against two Gram-positive (Staphylococcus aureus and Bacillus subtilis) and two Gram-negative (Escherichia coli and Xanthomonas campestris) bacterial strains. The complexes interact strongly with calf-thymus DNA via intercalation, as deduced by diverse techniques and via the determination of their binding constants. Complexes interact reversibly with bovine and human serum albumin. Complementary insights into their possible mechanisms of bioactivity at the molecular level were provided by molecular docking calculations, exploring in silico their ability to bind to calf-thymus DNA, Escherichia coli and Staphylococcus aureus DNA-gyrase, 5-lipoxygenase, and membrane transport lipid protein 5-lipoxygenase-activating protein, contributing to the understanding of the role complexes 1–5 can play both as antioxidant and antibacterial agents. Furthermore, in silico predictive tools have been employed to study the chemical reactivity, molecular properties and drug-likeness of the complexes, and also the drug-induced changes of gene expression profile (as protein- and mRNA-based prediction results), the sites of metabolism, the substrate/metabolite specificity, the cytotoxicity for cancer and non-cancer cell lines, the acute rat toxicity, the rodent organ-specific carcinogenicity, the anti-target interaction profiles, the environmental ecotoxicity, and finally the activity spectra profile of the compounds.
Collapse
|
9
|
de Souza MA, de Castro KK, Almeida-Neto FW, Bandeira PN, Ferreira MK, Marinho MM, da Rocha MN, de Brito DH, Mendes FRDS, Rodrigues TH, de Oliveira MR, de Menezes JE, Barreto AC, Marinho ES, de Lima-Neto P, dos Santos HS, Teixeira AM. Structural and spectroscopic analysis, ADMET study, and anxiolytic-like effect in adult zebrafish (Danio rerio) of 4′-[(1E,2E)-1-(2-(2′,4′-dinitrophenyl)hydrazone-3-(4-methoxyphenyl)allyl)aniline. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.132064] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
10
|
Ivanov SM, Lagunin AA, Filimonov DA, Poroikov VV. Relationships between the Structure and Severe Drug-Induced Liver Injury for Low, Medium, and High Doses of Drugs. Chem Res Toxicol 2022; 35:402-411. [PMID: 35172101 DOI: 10.1021/acs.chemrestox.1c00307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Assessment of structure-activity relationships (SARs) for predicting severe drug-induced liver injury (DILI) is essential since in vivo and in vitro preclinical methods cannot detect many druglike compounds disrupting liver functions. To date, plenty of SAR models for the prediction of DILI have been developed; however, none of them considered the route of drug administration and daily dose, which may introduce significant bias into prediction results. We have created a dataset of 617 drugs with parenteral and oral administration routes and consistent information on DILI severity. We have found a clear relationship between route, dose, and DILI severity. According to SAR, nearly 40% of moderate- and non-DILI-causing drugs would cause severe DILI if they were administered at high oral doses. We have proposed the following approach to predict severe DILI. New compounds recommended to be used at low oral doses (<∼10 mg daily), or parenterally, can be considered not causing severe DILI. DILI for compounds administered at medium oral doses (∼10-100 mg daily; 22.2% of drugs under consideration) can be considered unpredictable because reasonable SAR models were not obtained due to the small size and heterogeneity of the corresponding dataset. The DILI potential of the compounds recommended to be used at high oral doses (more than ∼100 mg daily) can be estimated using SAR modeling. The balanced accuracy of the approach calculated by a 10-fold cross-validation procedure is 0.803. The developed approach can be used to estimate severe DILI for druglike compounds proposed to use at low and high oral doses or parenterally at the early stages of drug development.
Collapse
Affiliation(s)
- Sergey M Ivanov
- Institute of Biomedical Chemistry, Pogodinskaya Str., 10/8, Moscow 119121, Russia.,Pirogov Russian National Research Medical University, Ostrovityanova Str., 1, Moscow 117997, Russia
| | - Alexey A Lagunin
- Institute of Biomedical Chemistry, Pogodinskaya Str., 10/8, Moscow 119121, Russia.,Pirogov Russian National Research Medical University, Ostrovityanova Str., 1, Moscow 117997, Russia
| | - Dmitry A Filimonov
- Institute of Biomedical Chemistry, Pogodinskaya Str., 10/8, Moscow 119121, Russia
| | - Vladimir V Poroikov
- Institute of Biomedical Chemistry, Pogodinskaya Str., 10/8, Moscow 119121, Russia
| |
Collapse
|
11
|
Ervina M, Pratama MRF, Poerwono H, Ekowati J, Widyowati R, Matsunami K, Sukardiman. In silico estrogen receptor alpha antagonist studies and toxicity prediction of Melia azedarach leaves bioactive ethyl acetate fraction. J Adv Pharm Technol Res 2021; 12:236-241. [PMID: 34345601 PMCID: PMC8300330 DOI: 10.4103/japtr.japtr_198_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/03/2021] [Accepted: 04/21/2021] [Indexed: 11/22/2022] Open
Abstract
The estrogen hormone dependent accounts for a major cause in the incidence of women breast cancer. Thus, their receptor, especially the estrogen receptor α (ER-α), is becoming a target in endocrine treatment. These ligand-inducible nuclear functions are regulated by an array of phytochemical and synthetic compounds, such as 17 β-estradiol and tamoxifen (4-hydroxytamoxifen [4OHT]). The Chinaberry (Melia azedarach) leaves are known naturally for relieving internal and external diseases. Previous studies revealed the potency of Melia's ethanolic extract and ethyl acetate fractions as anticancer; furthermore, this study aimed to resolve possible ER-α antagonist's mechanism and safety from M. azedarach leaves ethyl acetate fraction contents. Melia's phytochemical content was analyzed with electrospray ionization liquid chromatography-mass spectrometry, while its ER-α antagonist's potency was investigated by in silico. The computational docking was used to 3ERT (a human ER-α-4OHT binding domain complex) with Autodock Vina and related programs. The results presented Energy binding (ΔG) of Melia's quercetin 3-O-(2'',6''-digalloyl)-β-D-galactopyranoside was similar to 4OHT, and lower than its agonist 17 β-estradiol. Furthermore, the toxicity prediction of these compounds were revealed safer than 4OHT. The Melia's leaves ethyl acetate fraction, therefore, is a potential pharmacological material for further studies.
Collapse
Affiliation(s)
- Martha Ervina
- Doctoral Program of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Palangka Raya, Indonesia.,Department of Pharmaceutical Biology, Faculty of Pharmacy, Widya Mandala Catholic University, Surabaya, Indonesia
| | - Mohammad Rizki Fadhil Pratama
- Doctoral Program of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Palangka Raya, Indonesia.,Department of Pharmacy, Faculty of Health Sciences, Universitas Muhammadiyah Palangkaraya, Palangka Raya, Indonesia
| | - Hadi Poerwono
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Indonesia
| | - Juni Ekowati
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Indonesia
| | - Retno Widyowati
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Indonesia
| | - Katsuyoshi Matsunami
- Department of Pharmacognosy, Graduate School of Biomedical and Health Sciences, Hiroshima University, Higashihiroshima, Japan
| | - Sukardiman
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Indonesia
| |
Collapse
|
12
|
QSAR Assessing the Efficiency of Antioxidants in the Termination of Radical-Chain Oxidation Processes of Organic Compounds. Molecules 2021; 26:molecules26020421. [PMID: 33466934 PMCID: PMC7830365 DOI: 10.3390/molecules26020421] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/07/2021] [Accepted: 01/09/2021] [Indexed: 11/16/2022] Open
Abstract
Using the GUSAR 2013 program, the quantitative structure–antioxidant activity relationship has been studied for 74 phenols, aminophenols, aromatic amines and uracils having lgk7 = 0.01–6.65 (where k7 is the rate constant for the reaction of antioxidants with peroxyl radicals generated upon oxidation). Based on the atomic descriptors (Quantitative Neighborhood of Atoms (QNA) and Multilevel Neighborhoods of Atoms (MNA)) and molecular (topological length, topological volume and lipophilicity) descriptors, we have developed 9 statistically significant QSAR consensus models that demonstrate high accuracy in predicting the lgk7 values for the compounds of training sets and appropriately predict lgk7 for the test samples. Moderate predictive power of these models is demonstrated using metrics of two categories: (1) based on the determination coefficients R2 (R2TSi, R20, Q2(F1), Q2(F2), RmTSi2¯) and based on the concordance correlation coefficient (CCC)); or (2) based on the prediction lgk7 errors (root mean square error (RMSEP), mean absolute error (MAE) and standard deviation (S.D.)) The RBF-SCR method has been used for selecting the descriptors. Our theoretical prognosis of the lgk7 for 8-PPDA, a known antioxidant, based on the consensus models well agrees with the experimental value measure in the present work. Thus, the algorithms for calculating the descriptors implemented in the GUSAR 2013 program allow simulating kinetic parameters of the reactions underling the liquid-phase oxidation of hydrocarbons.
Collapse
|
13
|
Martynova YZ, Khairullina VR, Nasretdinova RN, Garifullina GG, Mitsukova DS, Gerchikov AY, Mustafin AG. Determination of the chain termination rate constants of the radical chain oxidation of organic compounds on antioxidant molecules by the QSPR method. Russ Chem Bull 2020. [DOI: 10.1007/s11172-020-2948-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
14
|
Poroikov VV. Computer-Aided Drug Design: from Discovery of Novel Pharmaceutical Agents to Systems Pharmacology. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES B: BIOMEDICAL CHEMISTRY 2020. [DOI: 10.1134/s1990750820030117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
15
|
Poroikov VV. [Computer-aided drug design: from discovery of novel pharmaceutical agents to systems pharmacology]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2020; 66:30-41. [PMID: 32116224 DOI: 10.18097/pbmc20206601030] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
New drug discovery is based on the analysis of public information about the mechanisms of the disease, molecular targets, and ligands, which interaction with the target could lead to the normalization of the pathological process. The available data on diseases, drugs, pharmacological effects, molecular targets, and drug-like substances, taking into account the combinatorics of the associative relations between them, correspond to the Big Data. To analyze such data, the application of computer-aided drug design methods is necessary. An overview of the studies in this area performed by the Laboratory for Structure-Function Based Drug Design of IBMC is presented. We have developed the approaches to identifying promising pharmacological targets, predicting several thousand types of biological activity based on the structural formula of the compound, analyzing protein-ligand interactions based on assessing local similarity of amino acid sequences, identifying likely molecular mechanisms of side effects of drugs, calculating the integral toxicity of drugs taking into account their metabolism, have been developed in the human body, predicting sustainable and sensitive options strains and evaluating the effectiveness of combinations of antiretroviral drugs in patients, taking into account the molecular genetic characteristics of the clinical isolates of HIV-1. Our computer programs are implemented as the web-services freely available on the Internet, which are used by thousands of researchers from many countries of the world to select the most promising substances for the synthesis and determine the priority areas for experimental testing of their biological activity.
Collapse
Affiliation(s)
- V V Poroikov
- Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
16
|
Elshakre ME, Noamaan MA, Moustafa H, Butt H. Density Functional Theory, Chemical Reactivity, Pharmacological Potential and Molecular Docking of Dihydrothiouracil-Indenopyridopyrimidines with Human-DNA Topoisomerase II. Int J Mol Sci 2020; 21:E1253. [PMID: 32070048 PMCID: PMC7072893 DOI: 10.3390/ijms21041253] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/25/2020] [Accepted: 01/27/2020] [Indexed: 01/31/2023] Open
Abstract
In this work, three computational methods (Hatree-Fock (HF), Møller-Plesset 2 (MP2), and Density Functional Theory (DFT)) using a variety of basis sets are used to determine the atomic and molecular properties of dihydrothiouracil-based indenopyridopyrimidine (TUDHIPP) derivatives. Reactivity descriptors of this system, including chemical potential (µ), chemical hardness (η), electrophilicity (ω), condensed Fukui function and dual descriptors are calculated at B3LYP/6-311++ G (d,p) to identify reactivity changes of these molecules in both gas and aqueous phases. We determined the molecular electrostatic surface potential (MESP) to determine the most active site in these molecules. Molecular docking study of TUDHIPP with topoisomerase II α and β is performed, predicting binding sites and binding energies with amino acids of both proteins. Docking studies of TUDHIPP versus etoposide suggest their potential as antitumor candidates. We have applied Lipinski, Veber's rules and analysis of the Golden triangle and structure activity/property relationship for a series of TUDHIPP derivatives indicate that the proposed compounds exhibit good oral bioavailability. The comparison of the drug likeness descriptors of TUDHIPP with those of etoposide, which is known to be an antitumor drug, indicates that TUDHIPP can be considered as an antitumor drug. The overall study indicates that TUDHIPP has comparable and even better descriptors than etoposide proposing that it can be as effective antitumor drug, especially 2H, 6H and 7H compounds.
Collapse
Affiliation(s)
- Mohamed E. Elshakre
- Chemistry Department, College of Science, Cairo University, Cairo 12613, Egypt;
| | - Mahmoud A. Noamaan
- Chemistry Department, College of Science, Cairo University, Cairo 12613, Egypt;
| | - Hussein Moustafa
- Chemistry Department, College of Science, Cairo University, Cairo 12613, Egypt;
| | - Haider Butt
- Department of Mechanical Engineering, Khalifa University, Abu Dhabi 127788, UAE;
| |
Collapse
|
17
|
Computer-aided prediction of biological activity spectra for organic compounds: the possibilities and limitations. Russ Chem Bull 2020. [DOI: 10.1007/s11172-019-2683-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
18
|
Yang ZY, He JH, Lu AP, Hou TJ, Cao DS. Application of Negative Design To Design a More Desirable Virtual Screening Library. J Med Chem 2020; 63:4411-4429. [DOI: 10.1021/acs.jmedchem.9b01476] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Zi-Yi Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, P. R. China
| | - Jun-Hong He
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, P. R. China
| | - Ai-Ping Lu
- Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, P. R. China
| | - Ting-Jun Hou
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, P. R. China
| | - Dong-Sheng Cao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, P. R. China
- Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, P. R. China
| |
Collapse
|
19
|
|
20
|
Dmitriev AV, Filimonov DA, Rudik AV, Pogodin PV, Karasev DA, Lagunin AA, Poroikov VV. Drug-drug interaction prediction using PASS. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2019; 30:655-664. [PMID: 31482727 DOI: 10.1080/1062936x.2019.1653966] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 08/06/2019] [Indexed: 06/10/2023]
Abstract
Simultaneous use of the drugs may lead to undesirable Drug-Drug Interactions (DDIs) in the human body. Many DDIs are associated with changes in drug metabolism that performed by Drug-Metabolizing Enzymes (DMEs). In this case, DDI manifests itself as a result of the effect of one drug on the biotransformation of other drug(s), its slowing down (in the case of inhibiting DME) or acceleration (in case of induction of DME), which leads to a change in the pharmacological effect of the drugs combination. We used OpeRational ClassificAtion (ORCA) system for categorizing DDIs. ORCA divides DDIs into five classes: contraindicated (class 1), provisionally contraindicated (class 2), conditional (class 3), minimal risk (class 4), no interaction (class 5). We collected a training set consisting of several thousands of drug pairs. Algorithm of PASS program was used for the first, second and third classes DDI prediction. Chemical descriptors called PoSMNA (Pairs of Substances Multilevel Neighbourhoods of Atoms) were developed and implemented in PASS software to describe in a machine-readable format drug substances pairs instead of the single molecules. The average accuracy of DDI class prediction is about 0.84. A freely available web resource for DDI prediction was developed (http://way2drug.com/ddi/).
Collapse
Affiliation(s)
- A V Dmitriev
- Department for Bioinformatics, Institute of Biomedical Chemistry (IBMC), Moscow, Russia
| | - D A Filimonov
- Department for Bioinformatics, Institute of Biomedical Chemistry (IBMC), Moscow, Russia
| | - A V Rudik
- Department for Bioinformatics, Institute of Biomedical Chemistry (IBMC), Moscow, Russia
| | - P V Pogodin
- Department for Bioinformatics, Institute of Biomedical Chemistry (IBMC), Moscow, Russia
| | - D A Karasev
- Department for Bioinformatics, Institute of Biomedical Chemistry (IBMC), Moscow, Russia
| | - A A Lagunin
- Department for Bioinformatics, Institute of Biomedical Chemistry (IBMC), Moscow, Russia
- Medico-biological Faculty, Pirogov Russian National Research Medical University, Moscow, Russia
| | - V V Poroikov
- Department for Bioinformatics, Institute of Biomedical Chemistry (IBMC), Moscow, Russia
| |
Collapse
|
21
|
Martynova YZ, Khairullina VR, Gimadieva AR, Mustafin AG. [QSAR-modeling of desoxyuridine triphosphatase inhibitors in a series of some derivatives of uracil]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2019; 65:103-113. [PMID: 30950815 DOI: 10.18097/pbmc20196502103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Due to the widespread prevalence, deoxyuridine triphosphatase (UTPase) is considered by modern biochemists and physicians as a promising target for the development of drugs with a wide range of activities. The therapeutic effect of these drugs will be due to suppression of DNA biosynthesis in various viruses, bacteria and protozoa. In order to rationalize the search for new dUTPase inhibitors, domestic and foreign researchers are actively using the QSAR methodology at the selection stage of hit compounds. However, the practical application of this methodology is impossible without existence of valid QSAR models. With the use of the GUSAR 2013 program, a quantitative analysis of the relationship between the structure and efficacy of 135 dUTPase inhibitors based on uracil derivatives was performed in the IC50 range of 30¸185000 nmol/L. Six statistically significant valid consensus models, characterized by high descriptive ability and moderate prognostic ability on the structures of training and test samples, are constructed. To build valid QSAR models for dUTPase inhibitors can use QNA or MNA descriptors and their combinations in a consensus approach.
Collapse
|
22
|
Dmitriev AV, Lagunin AA, Karasev DА, Rudik AV, Pogodin PV, Filimonov DA, Poroikov VV. Prediction of Drug-Drug Interactions Related to Inhibition or Induction of Drug-Metabolizing Enzymes. Curr Top Med Chem 2019; 19:319-336. [PMID: 30674264 DOI: 10.2174/1568026619666190123160406] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/02/2019] [Accepted: 01/07/2019] [Indexed: 02/07/2023]
Abstract
Drug-drug interaction (DDI) is the phenomenon of alteration of the pharmacological activity of a drug(s) when another drug(s) is co-administered in cases of so-called polypharmacy. There are three types of DDIs: pharmacokinetic (PK), pharmacodynamic, and pharmaceutical. PK is the most frequent type of DDI, which often appears as a result of the inhibition or induction of drug-metabolising enzymes (DME). In this review, we summarise in silico methods that may be applied for the prediction of the inhibition or induction of DMEs and describe appropriate computational methods for DDI prediction, showing the current situation and perspectives of these approaches in medicinal and pharmaceutical chemistry. We review sources of information on DDI, which can be used in pharmaceutical investigations and medicinal practice and/or for the creation of computational models. The problem of the inaccuracy and redundancy of these data are discussed. We provide information on the state-of-the-art physiologically- based pharmacokinetic modelling (PBPK) approaches and DME-based in silico methods. In the section on ligand-based methods, we describe pharmacophore models, molecular field analysis, quantitative structure-activity relationships (QSAR), and similarity analysis applied to the prediction of DDI related to the inhibition or induction of DME. In conclusion, we discuss the problems of DDI severity assessment, mention factors that influence severity, and highlight the issues, perspectives and practical using of in silico methods.
Collapse
Affiliation(s)
| | - Alexey A Lagunin
- Institute of Biomedical Chemistry, Moscow, Russian Federation.,Pirogov Russian National Research Medical University, Moscow, RussiaN Federation
| | | | | | - Pavel V Pogodin
- Institute of Biomedical Chemistry, Moscow, Russian Federation
| | | | | |
Collapse
|
23
|
Lagunin AA, Geronikaki A, Eleftheriou P, Pogodin PV, Zakharov AV. Rational Use of Heterogeneous Data in Quantitative Structure-Activity Relationship (QSAR) Modeling of Cyclooxygenase/Lipoxygenase Inhibitors. J Chem Inf Model 2019; 59:713-730. [PMID: 30688458 DOI: 10.1021/acs.jcim.8b00617] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Numerous studies have been published in recent years with acceptable quantitative structure-activity relationship (QSAR) modeling based on heterogeneous data. In many cases, the training sets for QSAR modeling were constructed from compounds tested by different biological assays, contradicting the opinion that QSAR modeling should be based on the data measured by a single protocol. We attempted to develop approaches that help to determine how heterogeneous data should be used for the creation of QSAR models on the basis of different sets of compounds tested by different experimental methods for the same target and the same endpoint. To this end, more than 100 QSAR models for the IC50 values of ligands interacting with cyclooxygenase 1,2 (COX) and seed lipoxygenase (LOX), obtained from ChEMBL database were created using the GUSAR software. The QSAR models were tested on the external set, including 26 new thiazolidinone derivatives, which were experimentally tested for COX-1,2/LOX inhibition. The IC50 values of the derivatives varied from 89 μM to 26 μM for LOX, from 200 μM to 0.018 μM for COX-1, and from 210 μM to 1 μM for COX-2. This study showed that the accuracy of the models is dependent on the distribution of IC50 values of low activity compounds in the training sets. In the most cases, QSAR models created based on the combined training sets had advantages in comparison with QSAR models, based on a single publication. We introduced a new method of combination of quantitative data from different experimental studies based on the data of reference compounds, which was called "scaling".
Collapse
Affiliation(s)
- Alexey A Lagunin
- Pirogov Russian National Research Medical University , Ostrovitianov str. 1 , Moscow , 117997 , Russia
- Institute of Biomedical Chemistry , Pogodinskaya Str., 10/8 , Moscow , 119121 , Russia
| | - Athina Geronikaki
- School of Pharmacy , Aristotle University , Thessaloniki , 54124 , Greece
| | - Phaedra Eleftheriou
- School of Health and Medical Care , Alexander Technological Educational Institute of Thessaloniki , Thessaloniki , 57400 , Greece
| | - Pavel V Pogodin
- Institute of Biomedical Chemistry , Pogodinskaya Str., 10/8 , Moscow , 119121 , Russia
| | - Alexey V Zakharov
- National Center for Advancing Translational Sciences (NCATS) , National Institutes of Health , Rockville , Maryland 20850 , United States
| |
Collapse
|
24
|
Manganese coordination compounds of mefenamic acid: In vitro screening and in silico prediction of biological activity. J Inorg Biochem 2019; 190:1-14. [DOI: 10.1016/j.jinorgbio.2018.09.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 09/14/2018] [Accepted: 09/26/2018] [Indexed: 02/07/2023]
|
25
|
Martynova YZ, Khairullina VR, Biglova YN, Mustafin AG. Quantitative structure-property relationship modeling of the C 60 fullerene derivatives as electron acceptors of polymer solar cells: Elucidating the functional groups critical for device performance. J Mol Graph Model 2018; 88:49-61. [PMID: 30660983 DOI: 10.1016/j.jmgm.2018.12.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 11/30/2018] [Accepted: 12/26/2018] [Indexed: 10/27/2022]
Abstract
Using the GUSAR 2013 program, we have performed a quantitative analysis of the "structure-power conversion efficiency (PCE)" on the series of 100 methano[60]fullerenes previously tested as acceptor components of bulk-heterojunction polymer organic solar cells (PSCs) utilizing the same donor polymer, viz. poly(3-hexylthiophene). Based on the MNA and QNA descriptors and self-consistent regression implemented in the program, six statistically significant consensus models for predicting the PCE values of the methano[60]fullerene-based PSCs have been constructed. The structural fragments of the fullerene compounds leading to an increase in the device performances are determined. Based on these structural descriptors, we have designed the three methano[60]fullerenes included in the training sets and characterized by poor optoelectrical properties is performed. As a result, two new compounds with potentially moderate efficiency have been proposed. This result opens opportunities of using the GUSAR 2013 program for modeling of the "structure-PCE" relationship for diverse compounds (not only fullerene derivatives).
Collapse
Affiliation(s)
- Yuliya Z Martynova
- Bashkir State University, 32 Z. Validi Str., Ufa, 450076, Russian Federation
| | | | - Yulya N Biglova
- Bashkir State University, 32 Z. Validi Str., Ufa, 450076, Russian Federation
| | - Akhat G Mustafin
- Bashkir State University, 32 Z. Validi Str., Ufa, 450076, Russian Federation; Ufa Institute of Chemistry of Russian Academy of Sciences, 71 Prospect Oktyabrya, Ufa, 450054, Russian Federation
| |
Collapse
|
26
|
Lagunin AA, Romanova MA, Zadorozhny AD, Kurilenko NS, Shilov BV, Pogodin PV, Ivanov SM, Filimonov DA, Poroikov VV. Comparison of Quantitative and Qualitative (Q)SAR Models Created for the Prediction of K i and IC 50 Values of Antitarget Inhibitors. Front Pharmacol 2018; 9:1136. [PMID: 30364128 PMCID: PMC6192375 DOI: 10.3389/fphar.2018.01136] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/18/2018] [Indexed: 12/20/2022] Open
Abstract
Estimation of interaction of drug-like compounds with antitargets is important for the assessment of possible toxic effects during drug development. Publicly available online databases provide data on the experimental results of chemical interactions with antitargets, which can be used for the creation of (Q)SAR models. The structures and experimental Ki and IC50 values for compounds tested on the inhibition of 30 antitargets from the ChEMBL 20 database were used. Data sets with Ki and IC50 values including more than 100 compounds were created for each antitarget. The (Q)SAR models were created by GUSAR software using quantitative neighborhoods of atoms (QNA), multilevel neighborhoods of atoms (MNA) descriptors, and self-consistent regression. The accuracy of (Q)SAR models was validated by the fivefold cross-validation procedure. The balanced accuracy was higher for qualitative SAR models (0.80 and 0.81 for Ki and IC50 values, respectively) than for quantitative QSAR models (0.73 and 0.76 for Ki and IC50 values, respectively). In most cases, sensitivity was higher for SAR models than for QSAR models, but specificity was higher for QSAR models. The mean R 2 and RMSE were 0.64 and 0.77 for Ki values and 0.59 and 0.73 for IC50 values, respectively. The number of compounds falling within the applicability domain was higher for SAR models than for the test sets.
Collapse
Affiliation(s)
- Alexey A. Lagunin
- Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow, Russia
- Department of Bioinformatics, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Maria A. Romanova
- Department of Bioinformatics, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Anton D. Zadorozhny
- Department of Bioinformatics, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Natalia S. Kurilenko
- Department of Bioinformatics, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Boris V. Shilov
- Department of Bioinformatics, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Pavel V. Pogodin
- Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow, Russia
| | - Sergey M. Ivanov
- Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow, Russia
- Department of Bioinformatics, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Dmitry A. Filimonov
- Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow, Russia
| | | |
Collapse
|
27
|
Khairullina VR, Gimadieva AR, Gerchikov AY, Mustafin AG, Zarudii FS. Quantitative structure-activity relationship of the thymidylate synthase inhibitors of Mus musculus in the series of quinazolin-4-one and quinazolin-4-imine derivatives. J Mol Graph Model 2018; 85:198-211. [PMID: 30227365 DOI: 10.1016/j.jmgm.2018.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/19/2018] [Accepted: 09/02/2018] [Indexed: 02/03/2023]
Abstract
A quantitative structure-activity relationship analysis of the 2-methylquinazolin-4-one and quinazolin-4-imine derivatives, well-known antifolate thymidylate synthase (TYMS) inhibitors, has been performed in the range IC50 = 0.4÷380000.0 nmoL/L using the GUSAR 2013 program. Based on the MNA and QNA descriptors using the self-consistent regression, 6 statistically significant consensus models for predicting the IC50 numerical values have been constructed. These models demonstrate high and moderate prognostic accuracies for the training and external validation test sets, respectively. The molecular fragments of TYMS inhibitors regulating their antitumor activity are identified. The obtained data open opportunities for developing novel promising inhibitors of TYMS.
Collapse
Affiliation(s)
| | - Alfiya R Gimadieva
- Ufa Institute of Chemistry - Subdivision of the Ufa Federal Research Centre of the Russian Academy of Sciences, 71 prospect Oktyabrya, Ufa, 450054, Russian Federation
| | | | - Akhat G Mustafin
- Bashkir State University, 32 Z. Validi str., Ufa, 450076, Russian Federation; Ufa Institute of Chemistry - Subdivision of the Ufa Federal Research Centre of the Russian Academy of Sciences, 71 prospect Oktyabrya, Ufa, 450054, Russian Federation
| | - Felix S Zarudii
- Bashkir State Medical University, 3 Lenina str, Ufa, 450000, Russian Federation
| |
Collapse
|
28
|
Hou TY, Weng CF, Leong MK. Insight Analysis of Promiscuous Estrogen Receptor α-Ligand Binding by a Novel Machine Learning Scheme. Chem Res Toxicol 2018; 31:799-813. [PMID: 30019586 DOI: 10.1021/acs.chemrestox.8b00130] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Estrogen receptor α (ERα) plays a significant role in occurrence of breast cancer and may cause various adverse side-effects when ERα is an off-target protein. A theoretical model was derived to predict the binding affinity of ERα using the pharmacophore ensemble/support vector machine (PhE/SVM) scheme to consider the promiscuous characteristic of ERα. The estimations by PhE/SVM were discovered to be in good agreement with the observed values for those training molecules ( n = 31, r2 = 0.80, qCV2 = 0.77, RMSE = 0.57, s = 0.58), test molecules ( n = 179, q2 = 0.91-0.96, RMSE = 0.33, s = 0.26) and outliers ( n = 15, q2 = 0.80-0.86, RMSE = 0.56, s = 0.49). When subjected to various statistical validations, the PhE/SVM model consistently fulfilled the strictest criteria. A mock test also asserted its predictivity. When compared with crystal structures, the calculated results are consistent with the reported ERα-ligand co-complex structure, and the plasticity nature of ERα is also disclosed. Consequently, this precise, fast, and robust model can be adopted to predict ERα-ligand binding affinities and to design safer non-ERα-targeted pharmaceuticals in the process of drug discovery and development.
Collapse
|
29
|
Capuzzi SJ, Sun W, Muratov EN, Martínez-Romero C, He S, Zhu W, Li H, Tawa G, Fisher EG, Xu M, Shinn P, Qiu X, García-Sastre A, Zheng W, Tropsha A. Computer-Aided Discovery and Characterization of Novel Ebola Virus Inhibitors. J Med Chem 2018; 61:3582-3594. [PMID: 29624387 DOI: 10.1021/acs.jmedchem.8b00035] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The Ebola virus (EBOV) causes severe human infection that lacks effective treatment. A recent screen identified a series of compounds that block EBOV-like particle entry into human cells. Using data from this screen, quantitative structure-activity relationship models were built and employed for virtual screening of a ∼17 million compound library. Experimental testing of 102 hits yielded 14 compounds with IC50 values under 10 μM, including several sub-micromolar inhibitors, and more than 10-fold selectivity against host cytotoxicity. These confirmed hits include FDA-approved drugs and clinical candidates with non-antiviral indications, as well as compounds with novel scaffolds and no previously known bioactivity. Five selected hits inhibited BSL-4 live-EBOV infection in a dose-dependent manner, including vindesine (0.34 μM). Additional studies of these novel anti-EBOV compounds revealed their mechanisms of action, including the inhibition of NPC1 protein, cathepsin B/L, and lysosomal function. Compounds identified in this study are among the most potent and well-characterized anti-EBOV inhibitors reported to date.
Collapse
Affiliation(s)
- Stephen J Capuzzi
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry , UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States
| | - Wei Sun
- National Center for Advancing Translational Sciences , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Eugene N Muratov
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry , UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States.,Department of Chemical Technology , Odessa National Polytechnic University , Odessa 65000 , Ukraine
| | - Carles Martínez-Romero
- Department of Microbiology , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States.,Global Health and Emerging Pathogens Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Shihua He
- Special Pathogens Program, National Microbiology Laboratory , Public Health Agency of Canada , 1015 Arlington Street , Winnipeg , Manitoba R3E 3R2 , Canada
| | - Wenjun Zhu
- Special Pathogens Program, National Microbiology Laboratory , Public Health Agency of Canada , 1015 Arlington Street , Winnipeg , Manitoba R3E 3R2 , Canada.,Department of Medical Microbiology , University of Manitoba , 745 Bannatyne Avenue , Winnipeg , Manitoba R3E 0J9 , Canada
| | - Hao Li
- National Center for Advancing Translational Sciences , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Gregory Tawa
- National Center for Advancing Translational Sciences , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Ethan G Fisher
- National Center for Advancing Translational Sciences , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Miao Xu
- National Center for Advancing Translational Sciences , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Paul Shinn
- National Center for Advancing Translational Sciences , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Xiangguo Qiu
- Special Pathogens Program, National Microbiology Laboratory , Public Health Agency of Canada , 1015 Arlington Street , Winnipeg , Manitoba R3E 3R2 , Canada.,Department of Medical Microbiology , University of Manitoba , 745 Bannatyne Avenue , Winnipeg , Manitoba R3E 0J9 , Canada
| | - Adolfo García-Sastre
- Department of Microbiology , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States.,Global Health and Emerging Pathogens Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States.,Department of Medicine, Division of Infectious Diseases , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Wei Zheng
- National Center for Advancing Translational Sciences , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Alexander Tropsha
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry , UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States
| |
Collapse
|
30
|
Druzhilovskiy DS, Rudik AV, Filimonov DA, Gloriozova TA, Lagunin AA, Dmitriev AV, Pogodin PV, Dubovskaya VI, Ivanov SM, Tarasova OA, Bezhentsev VM, Murtazalieva KA, Semin MI, Maiorov IS, Gaur AS, Sastry GN, Poroikov VV. Computational platform Way2Drug: from the prediction of biological activity to drug repurposing. Russ Chem Bull 2018. [DOI: 10.1007/s11172-017-1954-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
31
|
Khairullina VR, Gerchikov AY, Lagunin AA, Zarudii FS. QSAR Modelling of Thymidylate Synthase Inhibitors in a Series of Quinazoline Derivatives. Pharm Chem J 2018. [DOI: 10.1007/s11094-018-1710-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
32
|
Filimonov D, Druzhilovskiy D, Lagunin A, Gloriozova T, Rudik A, Dmitriev A, Pogodin P, Poroikov V. Computer-aided prediction of biological activity spectra for chemical compounds: opportunities and limitation. ACTA ACUST UNITED AC 2018. [DOI: 10.18097/bmcrm00004] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
An essential characteristic of chemical compounds is their biological activity since its presence can become the basis for the use of the substance for therapeutic purposes, or, on the contrary, limit the possibilities of its practical application due to the manifestation of side action and toxic effects. Computer assessment of the biological activity spectra makes it possible to determine the most promising directions for the study of the pharmacological action of particular substances, and to filter out potentially dangerous molecules at the early stages of research. For more than 25 years, we have been developing and improving the computer program PASS (Prediction of Activity Spectra for Substances), designed to predict the biological activity spectrum of substance based on the structural formula of its molecules. The prediction is carried out by the analysis of structure-activity relationships for the training set, which currently contains information on structures and known biological activities for more than one million molecules. The structure of the organic compound is represented in PASS using Multilevel Neighborhoods of Atoms descriptors; the activity prediction for new compounds is performed by the naive Bayes classifier and the structure-activity relationships determined by the analysis of the training set. We have created and improved both local versions of the PASS program and freely available web resources based on PASS (http://www.way2drug.com). They predict several thousand biological activities (pharmacological effects, molecular mechanisms of action, specific toxicity and adverse effects, interaction with the unwanted targets, metabolism and action on molecular transport), cytotoxicity for tumor and non-tumor cell lines, carcinogenicity, induced changes of gene expression profiles, metabolic sites of the major enzymes of the first and second phases of xenobiotics biotransformation, and belonging to substrates and/or metabolites of metabolic enzymes. The web resource Way2Drug is used by over 18,000 researchers from more than 90 countries around the world, which allowed them to obtain over 600,000 predictions and publish about 500 papers describing the obtained results. The analysis of the published works shows that in some cases the interpretation of the prediction results presented by the authors of these publications requires an adjustment. In this work, we provide the theoretical basis and consider, on particular examples, the opportunities and limitations of computer-aided prediction of biological activity spectra.
Collapse
Affiliation(s)
| | | | - A.A. Lagunin
- Institute of Biomedical Chemistry; Pirogov Russian National Research Medical University, Moscow, Russia
| | | | - A.V. Rudik
- Institute of Biomedical Chemistry, Moscow, Russia
| | | | - P.V. Pogodin
- Institute of Biomedical Chemistry, Moscow, Russia
| | | |
Collapse
|
33
|
|
34
|
Ozturk I, Yarar S, Banti C, Kourkoumelis N, Chrysouli M, Manoli M, Tasiopoulos A, Hadjikakou S. QSAR studies on antimony(III) halide complexes with N-substituted thiourea derivatives. Polyhedron 2017. [DOI: 10.1016/j.poly.2016.11.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
35
|
Ivanov S, Semin M, Lagunin A, Filimonov D, Poroikov V. In Silico Identification of Proteins Associated with Drug-induced Liver Injury Based on the Prediction of Drug-target Interactions. Mol Inform 2017; 36. [PMID: 28145637 DOI: 10.1002/minf.201600142] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 01/16/2017] [Indexed: 12/13/2022]
Abstract
Drug-induced liver injury (DILI) is the leading cause of acute liver failure as well as one of the major reasons for drug withdrawal from clinical trials and the market. Elucidation of molecular interactions associated with DILI may help to detect potentially hazardous pharmacological agents at the early stages of drug development. The purpose of our study is to investigate which interactions with specific human protein targets may cause DILI. Prediction of interactions with 1534 human proteins was performed for the dataset with information about 699 drugs, which were divided into three categories of DILI: severe (178 drugs), moderate (310 drugs) and without DILI (211 drugs). Based on the comparison of drug-target interactions predicted for different drugs' categories and interpretation of those results using clustering, Gene Ontology, pathway and gene expression analysis, we identified 61 protein targets associated with DILI. Most of the revealed proteins were linked with hepatocytes' death caused by disruption of vital cellular processes, as well as the emergence of inflammation in the liver. It was found that interaction of a drug with the identified targets is the essential molecular mechanism of the severe DILI for the most of the considered pharmaceuticals. Thus, pharmaceutical agents interacting with many of the identified targets may be considered as candidates for filtering out at the early stages of drug research.
Collapse
Affiliation(s)
- Sergey Ivanov
- Institute of Biomedical Chemistry 10 building 8, Pogodinskaya str., 119121, Moscow, Russia.,Pirogov Russian National Research Medical University, Medico-Biological Faculty 1, Ostrovitianova str., 117997, Moscow, Russia
| | - Maxim Semin
- Institute of Biomedical Chemistry 10 building 8, Pogodinskaya str., 119121, Moscow, Russia.,Pirogov Russian National Research Medical University, Medico-Biological Faculty 1, Ostrovitianova str., 117997, Moscow, Russia
| | - Alexey Lagunin
- Institute of Biomedical Chemistry 10 building 8, Pogodinskaya str., 119121, Moscow, Russia.,Pirogov Russian National Research Medical University, Medico-Biological Faculty 1, Ostrovitianova str., 117997, Moscow, Russia
| | - Dmitry Filimonov
- Institute of Biomedical Chemistry 10 building 8, Pogodinskaya str., 119121, Moscow, Russia
| | - Vladimir Poroikov
- Institute of Biomedical Chemistry 10 building 8, Pogodinskaya str., 119121, Moscow, Russia
| |
Collapse
|
36
|
Alves VM, Capuzzi SJ, Muratov E, Braga RC, Thornton T, Fourches D, Strickland J, Kleinstreuer N, Andrade CH, Tropsha A. QSAR models of human data can enrich or replace LLNA testing for human skin sensitization. GREEN CHEMISTRY : AN INTERNATIONAL JOURNAL AND GREEN CHEMISTRY RESOURCE : GC 2016; 18:6501-6515. [PMID: 28630595 PMCID: PMC5473635 DOI: 10.1039/c6gc01836j] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Skin sensitization is a major environmental and occupational health hazard. Although many chemicals have been evaluated in humans, there have been no efforts to model these data to date. We have compiled, curated, analyzed, and compared the available human and LLNA data. Using these data, we have developed reliable computational models and applied them for virtual screening of chemical libraries to identify putative skin sensitizers. The overall concordance between murine LLNA and human skin sensitization responses for a set of 135 unique chemicals was low (R = 28-43%), although several chemical classes had high concordance. We have succeeded to develop predictive QSAR models of all available human data with the external correct classification rate of 71%. A consensus model integrating concordant QSAR predictions and LLNA results afforded a higher CCR of 82% but at the expense of the reduced external dataset coverage (52%). We used the developed QSAR models for virtual screening of CosIng database and identified 1061 putative skin sensitizers; for seventeen of these compounds, we found published evidence of their skin sensitization effects. Models reported herein provide more accurate alternative to LLNA testing for human skin sensitization assessment across diverse chemical data. In addition, they can also be used to guide the structural optimization of toxic compounds to reduce their skin sensitization potential.
Collapse
Affiliation(s)
- Vinicius M. Alves
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
- Laboratory for Molecular Modeling and Design, Faculty of Pharmacy, Federal University of Goias, Goiania, GO, 74605-170, Brazil
| | - Stephen J. Capuzzi
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Eugene Muratov
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
- Department of Chemical Technology, Odessa National Polytechnic University, Odessa, 65000, Ukraine
| | - Rodolpho C. Braga
- Laboratory for Molecular Modeling and Design, Faculty of Pharmacy, Federal University of Goias, Goiania, GO, 74605-170, Brazil
| | - Thomas Thornton
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Denis Fourches
- Department of Chemistry, Bioinformatics Research Center, North Carolina State University, Raleigh, NC, 27695, USA
| | - Judy Strickland
- Integrated Laboratory Systems, Inc., P.O. Box 13501, Research Triangle Park, NC, 27709, USA
| | - Nicole Kleinstreuer
- National Institutes of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - Carolina H. Andrade
- Laboratory for Molecular Modeling and Design, Faculty of Pharmacy, Federal University of Goias, Goiania, GO, 74605-170, Brazil
| | - Alexander Tropsha
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| |
Collapse
|
37
|
Alves V, Muratov E, Capuzzi S, Politi R, Low Y, Braga R, Zakharov AV, Sedykh A, Mokshyna E, Farag S, Andrade C, Kuz'min V, Fourches D, Tropsha A. Alarms about structural alerts. GREEN CHEMISTRY : AN INTERNATIONAL JOURNAL AND GREEN CHEMISTRY RESOURCE : GC 2016; 18:4348-4360. [PMID: 28503093 PMCID: PMC5423727 DOI: 10.1039/c6gc01492e] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Structural alerts are widely accepted in chemical toxicology and regulatory decision support as a simple and transparent means to flag potential chemical hazards or group compounds into categories for read-across. However, there has been a growing concern that alerts disproportionally flag too many chemicals as toxic, which questions their reliability as toxicity markers. Conversely, the rigorously developed and properly validated statistical QSAR models can accurately and reliably predict the toxicity of a chemical; however, their use in regulatory toxicology has been hampered by the lack of transparency and interpretability. We demonstrate that contrary to the common perception of QSAR models as "black boxes" they can be used to identify statistically significant chemical substructures (QSAR-based alerts) that influence toxicity. We show through several case studies, however, that the mere presence of structural alerts in a chemical, irrespective of the derivation method (expert-based or QSAR-based), should be perceived only as hypotheses of possible toxicological effect. We propose a new approach that synergistically integrates structural alerts and rigorously validated QSAR models for a more transparent and accurate safety assessment of new chemicals.
Collapse
Affiliation(s)
- Vinicius Alves
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
- Laboratory for Molecular Modeling and Design, Department of Pharmacy, Federal University of Goias, Goiania, GO, 74605-170, Brazil
| | - Eugene Muratov
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
- Department of Chemical Technology, Odessa National Polytechnic University, Odessa, 65000, Ukraine
| | - Stephen Capuzzi
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Regina Politi
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Yen Low
- Netflix, San Francisco, CA 94123, USA
| | - Rodolpho Braga
- Laboratory for Molecular Modeling and Design, Department of Pharmacy, Federal University of Goias, Goiania, GO, 74605-170, Brazil
| | - Alexey V. Zakharov
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Rockville, MD 20850, USA
| | | | - Elena Mokshyna
- Laboratory of Theoretical Chemistry, A.V. Bogatsky Physical-Chemical Institute NAS of Ukraine, Odessa, 65080, Ukraine
| | - Sherif Farag
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Carolina Andrade
- Laboratory for Molecular Modeling and Design, Department of Pharmacy, Federal University of Goias, Goiania, GO, 74605-170, Brazil
| | - Victor Kuz'min
- Laboratory of Theoretical Chemistry, A.V. Bogatsky Physical-Chemical Institute NAS of Ukraine, Odessa, 65080, Ukraine
| | - Denis Fourches
- Department of Chemistry and Bioinformatics Research Center, North Carolina State University, Raleigh, NC, 27695, USA
| | - Alexander Tropsha
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| |
Collapse
|
38
|
Zakharov AV, Varlamova EV, Lagunin AA, Dmitriev AV, Muratov EN, Fourches D, Kuz'min VE, Poroikov VV, Tropsha A, Nicklaus MC. QSAR Modeling and Prediction of Drug-Drug Interactions. Mol Pharm 2016; 13:545-56. [PMID: 26669717 DOI: 10.1021/acs.molpharmaceut.5b00762] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Severe adverse drug reactions (ADRs) are the fourth leading cause of fatality in the U.S. with more than 100,000 deaths per year. As up to 30% of all ADRs are believed to be caused by drug-drug interactions (DDIs), typically mediated by cytochrome P450s, possibilities to predict DDIs from existing knowledge are important. We collected data from public sources on 1485, 2628, 4371, and 27,966 possible DDIs mediated by four cytochrome P450 isoforms 1A2, 2C9, 2D6, and 3A4 for 55, 73, 94, and 237 drugs, respectively. For each of these data sets, we developed and validated QSAR models for the prediction of DDIs. As a unique feature of our approach, the interacting drug pairs were represented as binary chemical mixtures in a 1:1 ratio. We used two types of chemical descriptors: quantitative neighborhoods of atoms (QNA) and simplex descriptors. Radial basis functions with self-consistent regression (RBF-SCR) and random forest (RF) were utilized to build QSAR models predicting the likelihood of DDIs for any pair of drug molecules. Our models showed balanced accuracy of 72-79% for the external test sets with a coverage of 81.36-100% when a conservative threshold for the model's applicability domain was applied. We generated virtually all possible binary combinations of marketed drugs and employed our models to identify drug pairs predicted to be instances of DDI. More than 4500 of these predicted DDIs that were not found in our training sets were confirmed by data from the DrugBank database.
Collapse
Affiliation(s)
- Alexey V Zakharov
- Computer-Aided Drug Design Group, Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, DHHS, NCI-Frederick , 376 Boyles Street, Frederick, Maryland 21702, United States
| | - Ekaterina V Varlamova
- Department of Molecular Structure and Cheminformatics, A.V. Bogatsky Physical Chemical Institute, National Academy of Sciences of Ukraine , Lustdorfskaya Doroga 86, Odessa 65080, Ukraine.,Chemical-Technological Department, Odessa National Polytechnic University , 1 Shevchenko Ave, Odessa 65000, Ukraine
| | - Alexey A Lagunin
- Institute of Biochemical Chemistry , 10/8, Pogodinskaya street, 119121 Moscow, Russia.,Medico-Biological Department, Pirogov Russian National Research Medical University , Ostrovitianov str. 1, Moscow 117997, Russia
| | - Alexander V Dmitriev
- Institute of Biochemical Chemistry , 10/8, Pogodinskaya street, 119121 Moscow, Russia
| | - Eugene N Muratov
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina , Beard Hall 301, CB#7568, Chapel Hill, North Carolina 27599, United States
| | - Denis Fourches
- Department of Chemistry, Bioinformatics Research Center, North Carolina State University , Raleigh, North Carolina 27695, United States
| | - Victor E Kuz'min
- Department of Molecular Structure and Cheminformatics, A.V. Bogatsky Physical Chemical Institute, National Academy of Sciences of Ukraine , Lustdorfskaya Doroga 86, Odessa 65080, Ukraine
| | - Vladimir V Poroikov
- Institute of Biochemical Chemistry , 10/8, Pogodinskaya street, 119121 Moscow, Russia
| | - Alexander Tropsha
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina , Beard Hall 301, CB#7568, Chapel Hill, North Carolina 27599, United States
| | - Marc C Nicklaus
- Computer-Aided Drug Design Group, Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, DHHS, NCI-Frederick , 376 Boyles Street, Frederick, Maryland 21702, United States
| |
Collapse
|
39
|
Joy A, Balaji S. Drug-likeness of Phytic Acid and Its Analogues. Open Microbiol J 2015; 9:141-9. [PMID: 26668666 PMCID: PMC4676049 DOI: 10.2174/1874285801509010141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 04/18/2015] [Accepted: 04/21/2015] [Indexed: 11/24/2022] Open
Abstract
Inositol hexakisphosphate is known to be the phosphorous reserve in plants particularly in the seeds. Though it
has been known for its antinutrient properties for many years, recent research shed light to reveal it as a novel anticancer
agent. Hence the present study investigates the drug-likeness of phytic acid and its analogues through bioinformatics
methods. Two potential cancer drug targets such as mitogen activated kinase and inositol 1,4,5-triphosphate receptor are
included in the study. Out of 50 selected analogues of phytic acid, 42 structures interact well with the chosen drug targets.
The best interacting structures are 1-diphosinositol pentakisphosphate and 2,3,4,5,6-pentaphosphonooxycyclohexyl
dihydrogen phosphate. For both of these structures, the negative binding energy obtained was -49.5 KJ/mol; this affirms
the stability of the complex. ADME properties are also predicted to assess the drug-like properties of the compounds. The
structure activity relationship model is generated for 12 compounds with experimental IC50 values.
Collapse
Affiliation(s)
- Amitha Joy
- Department of Biotechnology, Sahrdaya College of Engineering and Technology, Kodakara-680684, Thrissur, India; ; R&D Centre, Bharathiar University, Coimbatore, Tamilnadu, 641046, India
| | - S Balaji
- Department of Biotechnology, Manipal Institute of Technology, Manipal 576104, Karnataka, India
| |
Collapse
|
40
|
Hadjikakou S, Ozturk I, Banti C, Kourkoumelis N, Hadjiliadis N. Recent advances on antimony(III/V) compounds with potential activity against tumor cells. J Inorg Biochem 2015; 153:293-305. [DOI: 10.1016/j.jinorgbio.2015.06.006] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 06/03/2015] [Accepted: 06/06/2015] [Indexed: 11/25/2022]
|
41
|
Khayrullina VR, Gerchikov AY, Lagunin AA, Zarudii FS. Quantitative analysis of structure-activity relationships of tetrahydro-2H-isoindole cyclooxygenase-2 inhibitors. BIOCHEMISTRY (MOSCOW) 2015; 80:74-86. [PMID: 25754042 DOI: 10.1134/s0006297915010095] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Using the GUSAR program, structure-activity relationships on inhibition of cyclooxygenase-2 (COX-2) catalytic activity were quantitatively analyzed for twenty-six derivatives of 4,5,6,7-tetrahydro-2H-isoindole, 2,3-dihydro-1H-pyrrolyzine, and benzothiophene in the concentration range of 0.6-700 nmol/liter IC50 values. Six statistically significant consensus QSAR models for prediction of IC50 values were designed based on MNA- and QNA-descriptors and their combinations. These models demonstrated high accuracy in the prediction of IC50 values for structures of both training and test sets. Structural fragments of the COX-2 inhibitors capable of strengthening or weakening the desired property were determined using the same program. This information can be taken into consideration on molecular design of new COX-2 inhibitors. It was shown that in most cases, the influence of structural fragments on the inhibitory activity of the studied compounds revealed with the GUSAR program coincided with the results of expert evaluation of their effects based on known experimental data, and this can be used for optimization of structures to change the value of their biological activity.
Collapse
Affiliation(s)
- V R Khayrullina
- Bashkir State University, Faculty of Chemistry, Ufa, 450076, Russia.
| | | | | | | |
Collapse
|
42
|
Modelling of compound combination effects and applications to efficacy and toxicity: state-of-the-art, challenges and perspectives. Drug Discov Today 2015; 21:225-38. [PMID: 26360051 DOI: 10.1016/j.drudis.2015.09.003] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 07/30/2015] [Accepted: 09/01/2015] [Indexed: 01/18/2023]
Abstract
The development of treatments involving combinations of drugs is a promising approach towards combating complex or multifactorial disorders. However, the large number of compound combinations that can be generated, even from small compound collections, means that exhaustive experimental testing is infeasible. The ability to predict the behaviour of compound combinations in biological systems, whittling down the number of combinations to be tested, is therefore crucial. Here, we review the current state-of-the-art in the field of compound combination modelling, with the aim to support the development of approaches that, as we hope, will finally lead to an integration of chemical with systems-level biological information for predicting the effect of chemical mixtures.
Collapse
|
43
|
In silico assessment of adverse drug reactions and associated mechanisms. Drug Discov Today 2015; 21:58-71. [PMID: 26272036 DOI: 10.1016/j.drudis.2015.07.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 07/15/2015] [Accepted: 07/31/2015] [Indexed: 12/31/2022]
Abstract
During recent years, various in silico approaches have been developed to estimate chemical and biological drug features, for example chemical fragments, protein targets, pathways, among others, that correlate with adverse drug reactions (ADRs) and explain the associated mechanisms. These features have also been used for the creation of predictive models that enable estimation of ADRs during the early stages of drug development. In this review, we discuss various in silico approaches to predict these features for a certain drug, estimate correlations with ADRs, establish causal relationships between selected features and ADR mechanisms and create corresponding predictive models.
Collapse
|
44
|
Tarasova OA, Urusova AF, Filimonov DA, Nicklaus MC, Zakharov AV, Poroikov VV. QSAR Modeling Using Large-Scale Databases: Case Study for HIV-1 Reverse Transcriptase Inhibitors. J Chem Inf Model 2015; 55:1388-99. [PMID: 26046311 DOI: 10.1021/acs.jcim.5b00019] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Large-scale databases are important sources of training sets for various QSAR modeling approaches. Generally, these databases contain information extracted from different sources. This variety of sources can produce inconsistency in the data, defined as sometimes widely diverging activity results for the same compound against the same target. Because such inconsistency can reduce the accuracy of predictive models built from these data, we are addressing the question of how best to use data from publicly and commercially accessible databases to create accurate and predictive QSAR models. We investigate the suitability of commercially and publicly available databases to QSAR modeling of antiviral activity (HIV-1 reverse transcriptase (RT) inhibition). We present several methods for the creation of modeling (i.e., training and test) sets from two, either commercially or freely available, databases: Thomson Reuters Integrity and ChEMBL. We found that the typical predictivities of QSAR models obtained using these different modeling set compilation methods differ significantly from each other. The best results were obtained using training sets compiled for compounds tested using only one method and material (i.e., a specific type of biological assay). Compound sets aggregated by target only typically yielded poorly predictive models. We discuss the possibility of "mix-and-matching" assay data across aggregating databases such as ChEMBL and Integrity and their current severe limitations for this purpose. One of them is the general lack of complete and semantic/computer-parsable descriptions of assay methodology carried by these databases that would allow one to determine mix-and-matchability of result sets at the assay level.
Collapse
Affiliation(s)
- Olga A Tarasova
- †Institute of Biochemical Chemistry, 10-8, Pogodinskaya St., 119121, Moscow, Russia
| | - Aleksandra F Urusova
- †Institute of Biochemical Chemistry, 10-8, Pogodinskaya St., 119121, Moscow, Russia
| | - Dmitry A Filimonov
- †Institute of Biochemical Chemistry, 10-8, Pogodinskaya St., 119121, Moscow, Russia
| | - Marc C Nicklaus
- ‡CADD Group, Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, DHHS, NCI-Frederick, 376 Boyles St., Frederick, Maryland 21702, United States
| | - Alexey V Zakharov
- ‡CADD Group, Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, DHHS, NCI-Frederick, 376 Boyles St., Frederick, Maryland 21702, United States
| | - Vladimir V Poroikov
- †Institute of Biochemical Chemistry, 10-8, Pogodinskaya St., 119121, Moscow, Russia
| |
Collapse
|
45
|
Fedorova EV, Buryakina AV, Zakharov AV, Filimonov DA, Lagunin AA, Poroikov VV. Design, synthesis and pharmacological evaluation of novel vanadium-containing complexes as antidiabetic agents. PLoS One 2014; 9:e100386. [PMID: 25057899 PMCID: PMC4109918 DOI: 10.1371/journal.pone.0100386] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 05/27/2014] [Indexed: 11/19/2022] Open
Abstract
Based on the data about structure and antidiabetic activity of twenty seven vanadium and zinc coordination complexes collected from literature we developed QSAR models using the GUSAR program. These QSAR models were applied to 10 novel vanadium coordination complexes designed in silico in order to predict their hypoglycemic action. The five most promising substances with predicted potent hypoglycemic action were selected for chemical synthesis and pharmacological evaluation. The selected coordination vanadium complexes were synthesized and tested in vitro and in vivo for their hypoglycemic activities and acute rat toxicity. Estimation of acute rat toxicity of these five vanadium complexes was performed using a freely available web-resource (http://way2drug.com/GUSAR/acutoxpredict.html). It has shown that the selected compounds belong to the class of moderate toxic pharmaceutical agents, according to the scale of Hodge and Sterner. Comparison with the predicted data has demonstrated a reasonable correspondence between the experimental and predicted values of hypoglycemic activity and toxicity. Bis{tert-butyl[amino(imino)methyl]carbamato}oxovanadium (IV) and sodium(2,2'-Bipyridyl)oxo-diperoxovanadate(V) octahydrate were identified as the most potent hypoglycemic agents among the synthesized compounds.
Collapse
Affiliation(s)
- Elena V. Fedorova
- Saint-Petersburg State Chemical Pharmaceutical Academy, Ministry of Healthcare and Social Development of Russian Federation, Saint-Petersburg, Russian Federation
| | - Anna V. Buryakina
- Saint-Petersburg State Chemical Pharmaceutical Academy, Ministry of Healthcare and Social Development of Russian Federation, Saint-Petersburg, Russian Federation
| | - Alexey V. Zakharov
- National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States of America
- Orekhovich Institute of Biomedical Chemistry of Russian Academy of Medical Sciences, Moscow, Russian Federation
| | - Dmitry A. Filimonov
- Orekhovich Institute of Biomedical Chemistry of Russian Academy of Medical Sciences, Moscow, Russian Federation
| | - Alexey A. Lagunin
- Orekhovich Institute of Biomedical Chemistry of Russian Academy of Medical Sciences, Moscow, Russian Federation
| | - Vladimir V. Poroikov
- Orekhovich Institute of Biomedical Chemistry of Russian Academy of Medical Sciences, Moscow, Russian Federation
| |
Collapse
|
46
|
Ivanov SM, Lagunin AA, Pogodin PV, Filimonov DA, Poroikov VV. Identification of Drug-Induced Myocardial Infarction-Related Protein Targets through the Prediction of Drug–Target Interactions and Analysis of Biological Processes. Chem Res Toxicol 2014; 27:1263-81. [DOI: 10.1021/tx500147d] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Sergey M. Ivanov
- Orekhovich Institute
of Biomedical Chemistry of Russian Academy of Medical Sciences, 10, Pogodinskaya str., 119121 Moscow, Russia
| | - Alexey A. Lagunin
- Orekhovich Institute
of Biomedical Chemistry of Russian Academy of Medical Sciences, 10, Pogodinskaya str., 119121 Moscow, Russia
- Medico-biological
Faculty, Pirogov Russian National Research Medical University, 1,
Ostrovitianova str., 117997 Moscow, Russia
| | - Pavel V. Pogodin
- Orekhovich Institute
of Biomedical Chemistry of Russian Academy of Medical Sciences, 10, Pogodinskaya str., 119121 Moscow, Russia
- Medico-biological
Faculty, Pirogov Russian National Research Medical University, 1,
Ostrovitianova str., 117997 Moscow, Russia
| | - Dmitry A. Filimonov
- Orekhovich Institute
of Biomedical Chemistry of Russian Academy of Medical Sciences, 10, Pogodinskaya str., 119121 Moscow, Russia
| | - Vladimir V. Poroikov
- Orekhovich Institute
of Biomedical Chemistry of Russian Academy of Medical Sciences, 10, Pogodinskaya str., 119121 Moscow, Russia
- Medico-biological
Faculty, Pirogov Russian National Research Medical University, 1,
Ostrovitianova str., 117997 Moscow, Russia
| |
Collapse
|
47
|
Zakharov A, Peach ML, Sitzmann M, Nicklaus MC. A new approach to radial basis function approximation and its application to QSAR. J Chem Inf Model 2014; 54:713-9. [PMID: 24451033 PMCID: PMC3985791 DOI: 10.1021/ci400704f] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Indexed: 01/19/2023]
Abstract
We describe a novel approach to RBF approximation, which combines two new elements: (1) linear radial basis functions and (2) weighting the model by each descriptor's contribution. Linear radial basis functions allow one to achieve more accurate predictions for diverse data sets. Taking into account the contribution of each descriptor produces more accurate similarity values used for model development. The method was validated on 14 public data sets comprising nine physicochemical properties and five toxicity endpoints. We also compared the new method with five different QSAR methods implemented in the EPA T.E.S.T. program. Our approach, implemented in the program GUSAR, showed a reasonable accuracy of prediction and high coverage for all external test sets, providing more accurate prediction results than the comparison methods and even the consensus of these methods. Using our new method, we have created models for physicochemical and toxicity endpoints, which we have made freely available in the form of an online service at http://cactus.nci.nih.gov/chemical/apps/cap.
Collapse
Affiliation(s)
- Alexey
V. Zakharov
- CADD
Group, Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes
of Health, DHHS, NCI-Frederick, , 376 Boyles St., Frederick, Maryland 21702, United
States
| | - Megan L. Peach
- Basic
Science Program, Leidos Biomedical, Inc., Computer-Aided Drug Design Group, Chemical Biology Laboratory, Frederick
National Laboratory for Cancer Research, 376 Boyles St., Frederick, Maryland 21702, United States
| | - Markus Sitzmann
- CADD
Group, Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes
of Health, DHHS, NCI-Frederick, , 376 Boyles St., Frederick, Maryland 21702, United
States
| | - Marc C. Nicklaus
- CADD
Group, Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes
of Health, DHHS, NCI-Frederick, , 376 Boyles St., Frederick, Maryland 21702, United
States
| |
Collapse
|
48
|
Zakharov A, Peach ML, Sitzmann M, Nicklaus MC. QSAR modeling of imbalanced high-throughput screening data in PubChem. J Chem Inf Model 2014; 54:705-12. [PMID: 24524735 PMCID: PMC3985743 DOI: 10.1021/ci400737s] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Indexed: 01/19/2023]
Abstract
Many of the structures in PubChem are annotated with activities determined in high-throughput screening (HTS) assays. Because of the nature of these assays, the activity data are typically strongly imbalanced, with a small number of active compounds contrasting with a very large number of inactive compounds. We have used several such imbalanced PubChem HTS assays to test and develop strategies to efficiently build robust QSAR models from imbalanced data sets. Different descriptor types [Quantitative Neighborhoods of Atoms (QNA) and "biological" descriptors] were used to generate a variety of QSAR models in the program GUSAR. The models obtained were compared using external test and validation sets. We also report on our efforts to incorporate the most predictive of our models in the publicly available NCI/CADD Group Web services ( http://cactus.nci.nih.gov/chemical/apps/cap).
Collapse
Affiliation(s)
- Alexey
V. Zakharov
- CADD
Group, Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes
of Health, DHHS, NCI-Frederick, 376 Boyles St., Frederick, Maryland 21702, United
States
| | - Megan L. Peach
- Basic
Science Program, Leidos Biomedical, Inc., Computer-Aided Drug Design Group, Chemical Biology Laboratory, Frederick
National Laboratory for Cancer Research, 376 Boyles St., Frederick, Maryland 21702, United States
| | - Markus Sitzmann
- CADD
Group, Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes
of Health, DHHS, NCI-Frederick, 376 Boyles St., Frederick, Maryland 21702, United
States
| | - Marc C. Nicklaus
- CADD
Group, Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes
of Health, DHHS, NCI-Frederick, 376 Boyles St., Frederick, Maryland 21702, United
States
| |
Collapse
|
49
|
Abstract
Drug action can be rationalized as interaction of a molecule with proteins in a regulatory network of targets from a specific biological system. Both drug and side effects are often governed by interaction of the drug molecule with many, often unrelated, targets. Accordingly, arrays of protein–ligand interaction data from numerous in vitro profiling assays today provide growing evidence of polypharmacological drug interactions, even for marketed drugs. In vitro off-target profiling has therefore become an important tool in early drug discovery to learn about potential off-target liabilities, which are sometimes beneficial, but more often safety relevant. The rapidly developing field of in silico profiling approaches is complementing in vitro profiling. These approaches capitalize from large amounts of biochemical data from multiple sources to be exploited for optimizing undesirable side effects in pharmaceutical research. Therefore, current in silico profiling models are nowadays perceived as valuable tools in drug discovery, and promise a platform to support optimally informed decisions.
Collapse
|
50
|
Lagunin AA, Goel RK, Gawande DY, Pahwa P, Gloriozova TA, Dmitriev AV, Ivanov SM, Rudik AV, Konova VI, Pogodin PV, Druzhilovsky DS, Poroikov VV. Chemo- and bioinformatics resources for in silico drug discovery from medicinal plants beyond their traditional use: a critical review. Nat Prod Rep 2014; 31:1585-611. [DOI: 10.1039/c4np00068d] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
An overview of databases andin silicotools for discovery of the hidden therapeutic potential of medicinal plants.
Collapse
Affiliation(s)
- Alexey A. Lagunin
- Orekhovich Institute of Biomedical Chemistry of Rus. Acad. Med. Sci
- Moscow, Russia
- Russian National Research Medical University
- Medico-Biologic Faculty
- Moscow, Russia
| | - Rajesh K. Goel
- Department of Pharmaceutical Sciences and Drug Research
- Punjabi University
- Patiala-147002, India
| | - Dinesh Y. Gawande
- Department of Pharmaceutical Sciences and Drug Research
- Punjabi University
- Patiala-147002, India
| | - Priynka Pahwa
- Department of Pharmaceutical Sciences and Drug Research
- Punjabi University
- Patiala-147002, India
| | | | | | - Sergey M. Ivanov
- Orekhovich Institute of Biomedical Chemistry of Rus. Acad. Med. Sci
- Moscow, Russia
| | - Anastassia V. Rudik
- Orekhovich Institute of Biomedical Chemistry of Rus. Acad. Med. Sci
- Moscow, Russia
| | - Varvara I. Konova
- Orekhovich Institute of Biomedical Chemistry of Rus. Acad. Med. Sci
- Moscow, Russia
| | - Pavel V. Pogodin
- Orekhovich Institute of Biomedical Chemistry of Rus. Acad. Med. Sci
- Moscow, Russia
- Russian National Research Medical University
- Medico-Biologic Faculty
- Moscow, Russia
| | | | - Vladimir V. Poroikov
- Orekhovich Institute of Biomedical Chemistry of Rus. Acad. Med. Sci
- Moscow, Russia
- Russian National Research Medical University
- Medico-Biologic Faculty
- Moscow, Russia
| |
Collapse
|