330
Views
1
CrossRef citations to date
0
Altmetric
Research Articles

Naphthoquinones biflorin and bis-biflorin (Capraria biflora) as possible inhibitors of the fungus Candida auris polymerase: molecular docking, molecular dynamics, MM/GBSA calculations and in silico drug-likeness study

ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, , , , ORCID Icon, ORCID Icon & ORCID Icon show all
Pages 11564-11577 | Received 12 Apr 2022, Accepted 24 Dec 2022, Published online: 04 Jan 2023

References

  • Al-Jindan, R., & Al-Eraky, D. M. (2021). Two cases of the emerging Candida auris in a university hospital from Saudi Arabia. Saudi Journal of Medicine and Medical Sciences, 9(1), 71–74. https://doi.org/10.4103/sjmms.sjmms_449_19
  • Bai, Q., Tan, S., Xu, T., Liu, H., Huang, J., & Yao, X. (2021). MolAICal: A soft tool for 3D drug design of protein targets by artificial intelligence and classical algorithm. Briefings in Bioinformatics, 22(3), 1–12. https://doi.org/10.1093/bib/bbaa161
  • Bashir, Q., Li, Z., Li, H., LeMaster, D. M., & Hernández, G. (2020). Crystal structure and transient dimerization for the FKBP12 protein from the pathogenic fungus Candida auris. Biochemical and Biophysical Research Communications, 525(4), 1103–1108. https://doi.org/10.1016/j.bbrc.2020.03.059
  • Bergold, A. M., & Georgiadis, S. (2004). Novidades em fármacos antifúngico: Uma Revisão. Visão Acadêmica, 5(2), 159–172. https://doi.org/10.5380/acd.v5i2.562
  • Biovia. (2015). Dassault Systemes BIOVIA, Discovery Studio Modelling Environment, Release 4.5. In Accelrys Software Inc. (4.5).
  • Biovia, D. S. (2016). Discovery Studio Modeling Environment, Release 2017, San Diego. In Dassault Systèmes.
  • Cadnum, J. L., Shaikh, A. A., Piedrahita, C. T., Sankar, T., Jencson, A. L., Larkin, E. L., Ghannoum, M. A., & Donskey, C. J. (2017). Effectiveness of disinfectants against Candida auris and other candida species. Infection Control and Hospital Epidemiology, 38(10), 1240–1243. https://doi.org/10.1017/ice.2017.162
  • Cheatham, T. E., Miller, J. L., Fox, T., Darden, T. A., & Kollman, P. A. (1995). Molecular dynamics simulations on solvated biomolecular systems: The particle mesh ewald method leads to stable trajectories of DNA, RNA, and Proteins. Journal of the American Chemical Society, 117(14), 4193–4194. https://doi.org/10.1021/ja00119a045
  • Chen, F., Sun, H., Wang, J., Zhu, F., Liu, H., Wang, Z., Lei, T., Li, Y., & Hou, T. (2018). Assessing the performance of MM/PBSA and MM/GBSA methods. 8. Predicting binding free energies and poses of protein-RNA complexes. RNA (New York, N.Y.), 24(9), 1183–1194. https://doi.org/10.1261/rna.065896.118
  • da Silva, M. N., Ferreira, V. F., & de Souza, M. C. B. V. (2003). Um panorama atual da química e da farmacologia de naftoquinonas, com ênfase na beta-lapachona e derivados. Química Nova, 26(3), 407–416. https://doi.org/10.1590/S0100-40422003000300019
  • De Ruyck, J., Brysbaert, G., Blossey, R., & Lensink, M. F. (2016). Molecular docking as a popular tool in drug design, an in silico travel. Advances and Applications in Bioinformatics and Chemistry, 9, 1–11. https://doi.org/10.2147/AABC.S105289
  • De Vasconcellos, M. C., Bezerra, D. P., Fonseca, A. M., Araújo, A. J., Pessoa, C., Lemos, T. L. G., Costa-Lotufo, L. V., De Moraes, M. O., & Montenegro, R. C. (2011). The in-vitro and in-vivo inhibitory activity of biflorin in melanoma. Melanoma Research, 21(2), 106–114. https://doi.org/10.1097/CMR.0b013e328343ecc4
  • De Vasconcellos, M. C., Bezerra, D. P., Fonseca, A. M., Pereira, M. R. P., Lemos, T. L. G., Pessoa, O. D. L., Pessoa, C., De Moraes, M. O., Alves, A. P. N. N., & Costa-Lotufo, L. V. (2007). Antitumor activity of biflorin, an o-naphthoquinone isolated from Capraria biflora. Biological & Pharmaceutical Bulletin, 30(8), 1416–1421. https://doi.org/10.1248/bpb.30.1416
  • De Vasconcellos, M. C., Moura, D. J., Rosa, R. M., Machado, M. S., Guecheva, T. N., Villela, I., Immich, B. F., Montenegro, R. C., Fonseca, A. M., Lemos, T. L. G., Moraes, M. E. A., Saffi, J., Costa-Lotufo, L. V., Moraes, M. O., & Henriques, J. A. P. (2010). Evaluation of the cytotoxic and antimutagenic effects of biflorin, an antitumor 1,4 o-naphthoquinone isolated from Capraria biflora L. Archives of Toxicology, 84(10), 799–810. https://doi.org/10.1007/s00204-010-0567-z
  • Eyre, D. W., Sheppard, A. E., Madder, H., Moir, I., Moroney, R., Quan, T. P., Griffiths, D., George, S., Butcher, L., Morgan, M., Newnham, R., Sunderland, M., Clarke, T., Foster, D., Hoffman, P., Borman, A. M., Johnson, E. M., Moore, G., Brown, C. S., … Jeffery, K. J. M. (2018). A Candida auris outbreak and its control in an intensive care setting. The New England Journal of Medicine, 379(14), 1322–1331. https://doi.org/10.1056/NEJMoa1714373
  • Field, M. J., Albe, M., Bret, C., Proust-De Martin, F., & Thomas, A. (2000). The dynamo library for molecular simulations using hybrid quantum mechanical and molecular mechanical potentials. Journal of Computational Chemistry, 21(12), 1088–1100. https://doi.org/10.1002/1096-987X(200009)21:12 < 1088::AID-JCC5 > 3.0.CO;2-8
  • Fonseca, A., M., Pessoa, O. D. L., Silveira, E. R., Monte, F. J. Q., Braz-Filho, R., & Lemos, T. L. G. (2003). Total assignments of 1H and 13C NMR spectra of biflorin and bis-biflorin from Capraria biflora. Magnetic Resonance in Chemistry, 41(12), 1038–1040. https://doi.org/10.1002/mrc.1295
  • Frías-De-León, M. G., Hernández-Castro, R., Vite-Garín, T., Arenas, R., Bonifaz, A., Castañón-Olivares, L., Acosta-Altamirano, G., & Martínez-Herrera, E. (2020). Antifungal resistance in Candida auris: Molecular determinants. Antibiotics, 9(9), 1–16. https://doi.org/10.3390/antibiotics9090568
  • Genheden, S., & Ryde, U. (2015). The MM/PBSA and MM/GBSA methods to estimate ligand-binding affinities. Expert Opinion on Drug Discovery, 10(5), 449–461. https://doi.org/10.1517/17460441.2015.1032936
  • Giolo, M. P., & Svidzinski, T. I. E. (2010). Fisiopatogenia, epidemiologia e diagnóstico laboratorial da candidemia. Jornal Brasileiro De Patologia e Medicina Laboratorial, 46(3), 225–234. https://doi.org/10.1590/S1676-24442010000300009
  • Halgren, T. A. (1996). Merck molecular force field. I. Basis, form, scope, parameterization, and performance of MMFF94. Journal of Computational Chemistry, 17(5-6), 490–519. https://doi.org/10.1002/(SICI)1096-987X(199604)17:5/6 < 490::AID-JCC1 > 3.0.CO;2-P
  • Hanwell, M. D., Curtis, D. E., Lonie, D. C., Vandermeersch, T., Zurek, E., & Hutchison, G. R. (2012). Avogadro: An advanced semantic chemical editor, visualization, and analysis platform. Journal of Cheminformatics, 4(1), 17. 17. https://doi.org/10.1186/1758-2946-4-17
  • Hess, B., Bekker, H., Berendsen, H., J., C., & Fraaije, J. G. E. M. (1997). LINCS: A linear constraint solver for molecular simulations. Journal of Computational Chemistry, 18(12), 1463–1472. https://doi.org/10.1002/(SICI)1096-987X(199709)18:12 < 1463::AID-JCC4 > 3.0.CO;2-H
  • Hevener, K. E., Zhao, W., Ball, D. M., Babaoglu, K., Qi, J., White, S. W., & Lee, R. E. (2009). Validation of molecular docking programs for virtual screening against dihydropteroate synthase. Journal of Chemical Information and Modeling, 49(2), 444–460. https://doi.org/10.1021/ci800293n
  • Huang, H. J., Yu, H. W., Chen, C. Y., Hsu, C. H., Chen, H. Y., Lee, K. J., Tsai, F. J., Chen, C., & Y., C. (2010). Current developments of computer-aided drug design. Journal of the Taiwan Institute of Chemical Engineers, 41(6), 623–635. https://doi.org/10.1016/j.jtice.2010.03.017
  • Humphrey, W., Dalke, A., & Schulten, K. (1996). VMD: Visual molecular dynamics. Journal of Molecular Graphics, 14(1), 33–38. https://doi.org/10.1016/0263-7855(96)00018-5
  • Jeffery-Smith, A., Taori, S. K., Schelenz, S., Jeffery, K., Johnson, E. M., Borman, A., Manuel, R., & Brown, C. S. (2018). Candida auris: A review of the literature. Clinical Microbiology Reviews, 31(1), 1–18. https://doi.org/10.1128/CMR.00029-17
  • Jin, Z., Du, X., Xu, Y., Deng, Y., Liu, M., Zhao, Y., Zhang, B., Li, X., Zhang, L., Peng, C., Duan, Y., Yu, J., Wang, L., Yang, K., Liu, F., Jiang, R., Yang, X., You, T., Liu, X., … Yang, H. (2020). Structure of Mpro from SARS-CoV-2 and discovery of its inhibitors. Nature, 582(7811), 289–293. https://doi.org/10.1038/s41586-020-2223-y
  • Johnson, T. W., Dress, K. R., & Edwards, M. (2009). Using the Golden Triangle to optimize clearance and oral absorption. Bioorganic & Medicinal Chemistry Letters, 19(19), 5560–5564. https://doi.org/10.1016/j.bmcl.2009.08.045
  • Kelder, J., Grootenhuis, P. D. J., Bayada, D. M., Delbressine, L. P. C., & Ploemen, J.-P. (1999). Polar molecular surface as a dominating determinant for oral absorption and brain penetration of drugs. Pharmaceutical Research, 16(10), 1514–1519. https://doi.org/10.1023/A:1015040217741
  • Khan, R., Islam, B., Akram, M., Shakil, S., Ahmad, A., Ali, S. M., Siddiqui, M., & Khan, A. U. (2009). Antimicrobial activity of five herbal extracts against Multi Drug Resistant (MDR) strains of bacteria and fungus of clinical origin. Molecules, 14(2), 586–597. https://doi.org/10.3390/molecules14020586
  • Kim, S., Thiessen, P. A., Bolton, E. E., Chen, J., Fu, G., Gindulyte, A., Han, L., He, J., He, S., Shoemaker, B. A., Wang, J., Yu, B., Zhang, J., & Bryant, S. H. (2016). PubChem substance and compound databases. Nucleic Acids Research, 44(D1), D1202–D1213. https://doi.org/10.1093/nar/gkv951
  • Kordalewska, M., Zhao, Y., Lockhart, S. R., Chowdhary, A., Berrio, I., & Perlin, D. S. (2017). Rapid and accurate molecular identification of the emerging multidrug-resistant pathogen Candida auris. Journal of Clinical Microbiology, 55(8), 2445–2452. https://doi.org/10.1128/JCM.00630-17
  • Kumar, Y., Singh, H., & Patel, C. N. (2020). In silico prediction of potential inhibitors for the main protease of SARS-CoV-2 using molecular docking and dynamics simulation based drug-repurposing. Journal of Infection and Public Health, 13(9), 1210–1223. https://doi.org/10.1016/j.jiph.2020.06.016
  • Leite, J. D. P., Yamamoto, A., C. A., Martins, E. R., Teixeira, A. F. R., & Hahn, R. C. (2011). Leveduras do gênero Candida isoladas de sítios anatomicamente distintos de profissionais militares em Cuiabá (MT), Brasil. Anais Brasileiros De Dermatologia, 86(4), 675–680. https://doi.org/10.1590/S0365-05962011000400008
  • Lemos, T. L. G., Monte, F. J. Q., Santos, A. K. L., Fonseca, A. M., Santos, H. S., Oliveira, M. F., Costa, S. M. O., Pessoa, O. D. L., & Braz-Filho, R. (2007). Quinones from plants of northeastern Brazil: Structural diversity, chemical transformations, NMR data and biological activities. Natural Product Research, 21(6), 529–550. https://doi.org/10.1080/14786410601130604
  • Liu, F.-F., Dong, X.-Y., Wang, T., & Sun, Y. (2007). Rational design of peptide ligand for affinity chromatography of tissue-type plasminogen activator by the combination of docking and molecular dynamics simulations. Journal of Chromatography. A, 1175(2), 249–258. https://doi.org/10.1016/j.chroma.2007.10.074
  • Lockhart, S. R., Etienne, K. A., Vallabhaneni, S., Farooqi, J., Chowdhary, A., Govender, N. P., Colombo, A. L., Calvo, B., Cuomo, C., A., Desjardins, C., A., Berkow, E. L., Castanheira, M., Magobo, R. E., Jabeen, K., Asghar, R. J., Meis, J. F., Jackson, B., Chiller, T., & Litvintseva, A. P. (2017). Simultaneous emergence of multidrug-resistant Candida auris on 3 continents confirmed by whole-genome sequencing and epidemiological analyses. Clinical Infectious Diseases, 64(2), 134–140. https://doi.org/10.1093/cid/ciw691
  • Marinho, E. M., Batista de Andrade Neto, J., Silva, J., Rocha da Silva, C., Cavalcanti, B. C., Marinho, E. S., & Nobre Júnior, H. V. (2020). Virtual screening based on molecular docking of possible inhibitors of Covid-19 main protease. Microbial Pathogenesis, 148, 104365. https://doi.org/10.1016/j.micpath.2020.104365
  • Marques, B. S., Stetz, M. A., Jorge, C., Valentine, K. G., Wand, A. J., & Nucci, N. V. (2020). Protein conformational entropy is not slaved to water. Scientific Reports, 10(1), 1–8. https://doi.org/10.1038/s41598-020-74382-5
  • Martin, Y. C. (2005). A bioavailability score. Journal of Medicinal Chemistry, 48(9), 3164–3170. https://doi.org/10.1021/jm0492002
  • Martinez, R. (2006). Atualização no uso de agentes antifúngicos. Jornal Brasileiro De Pneumologia, 32(5), 449–460. https://doi.org/10.1590/S1806-37132006000500013
  • Mascoli, V., Liguori, N., Cupellini, L., Elias, E., Mennucci, B., & Croce, R. (2021). Uncovering the interactions driving carotenoid binding in light-harvesting complexes. Chemical Science, 12(14), 5113–5122. https://doi.org/10.1039/d1sc00071c
  • Menezes, E. A., Vasconcelos Júnior, A., de, A., Cunha, F. A., Cunha, M. C. d S. O., Braz, B. H. L., Capelo, L. G., & Silva, C. L. F. (2012). Identificação molecular e suscetibilidade antifúngica de Candida parapsilosis isoladas no Ceará, Brasil. Jornal Brasileiro De Patologia e Medicina Laboratorial, 48(6), 415–420. https://doi.org/10.1590/S1676-24442012000600005
  • Mohan, N., Suresh, C. H., Kumar, A., & Gadre, S. R. (2013). Molecular electrostatics for probing lone pair-π interactions. Physical Chemistry Chemical Physics, 15(42), 18401–18409. https://doi.org/10.1039/c3cp53379d
  • Nguyen, D. D., Xiao, T., Wang, M., & Wei, G. W. (2017). Rigidity strengthening: A mechanism for protein-ligand binding. Journal of Chemical Information and Modeling, 57(7), 1715–1721. https://doi.org/10.1021/acs.jcim.7b00226
  • Pelkonen, O., Turpeinen, M., & Raunio, H. (2011). In vivo-in vitro-in silico pharmacokinetic modelling in drug development. Clinical Pharmacokinetics, 50(8), 483–491. https://doi.org/10.2165/11592400-000000000-00000
  • Phillips, J. C., Braun, R., Wang, W., Gumbart, J., Tajkhorshid, E., Villa, E., Chipot, C., Skeel, R. D., Kalé, L., & Schulten, K. (2005). Scalable molecular dynamics with NAMD. Journal of Computational Chemistry, 26(16), 1781–1802. https://doi.org/10.1002/jcc.20289
  • Pozzatti, P., Loreto, É. S., Lopes, P. G. M., Athayde, M. L., Santurio, J. M., & Alves, S. H. (2010). Comparison of the susceptibilities of clinical isolates of Candida albicans and Candida dubliniensis to essential oils. Mycoses, 53(1), 12–15. https://doi.org/10.1111/j.1439-0507.2008.01643.x
  • Qin, X., Zhong, J., & Wang, Y. (2021). A mutant T1 lipase homology modeling, and its molecular docking and molecular dynamics simulation with fatty acids. Journal of Biotechnology, 337, 24–34. https://doi.org/10.1016/j.jbiotec.2021.06.024
  • Ragunathan, A., Malathi, K., & Anbarasu, A. (2018). MurB as a target in an alternative approach to tackle the Vibrio cholerae resistance using molecular docking and simulation study. Journal of Cellular Biochemistry, 119(2), 1726–1732. https://doi.org/10.1002/jcb.26333
  • Ritchie, T. J., Ertl, P., & Lewis, R. (2011). The graphical representation of ADME-related molecule properties for medicinal chemists. Drug Discovery Today, 16(1–2), 65–72. https://doi.org/10.1016/j.drudis.2010.11.002
  • Roe, D. R., & Brooks, B. R. (2020). A protocol for preparing explicitly solvated systems for stable molecular dynamics simulations. Journal of Chemical Physics, 153(5), 1–9. https://doi.org/10.1063/5.0013849
  • Ruba, S., Arooj, M., & Naz, D. G. (2014). In silico molecular docking studies and design of dengue virus inhibitors. IOSR Journal of Pharmacy and Biological Sciences, 9(2), 15–23. https://doi.org/10.9790/3008-09211523
  • Satoh, K., Makimura, K., Hasumi, Y., Nishiyama, Y., Uchida, K., & Yamaguchi, H. (2009). Candida auris sp. nov., a novel ascomycetous yeast isolated from the external ear canal of an inpatient in a Japanese hospital. Microbiology and Immunology, 53(1), 41–44. https://doi.org/10.1111/j.1348-0421.2008.00083.x
  • Sekizuka, T., Iguchi, S., Umeyama, T., Inamine, Y., Makimura, K., Kuroda, M., Miyazaki, Y., & Kikuchi, K. (2019). Clade II Candida auris possess genomic structural variations related to an ancestral strain. Plos One, 14(10), e0223433. https://doi.org/10.1371/journal.pone.0223433
  • Silva, T. L., Azevedo, M. L. S. G., Ferreira, F. R., Santos, D. C., Amatore, C., & Goulart, M. O. F. (2020). Quinone-based molecular electrochemistry and their contributions to medicinal chemistry: A look at the present and future. Current Opinion in Electrochemistry, 24, 79–87. https://doi.org/10.1016/j.coelec.2020.06.011
  • Sterling, T., & Irwin, J. J. (2015). ZINC 15 – Ligand discovery for everyone. Journal of Chemical Information and Modeling, 55(11), 2324–2337. https://doi.org/10.1021/acs.jcim.5b00559
  • Thön, M., Al-Abdallah, Q., Hortschansky, P., & Brakhage, A. A. (2007). The thioredoxin system of the filamentous fungus Aspergillus nidulans: Impact on development and oxidative stress response. Journal of Biological Chemistry, 282(37), 27259–27269. https://doi.org/10.1074/jbc.M704298200
  • Tortorano, A. M., Kibbler, C., Peman, J., Bernhardt, H., Klingspor, L., & Grillot, R. (2006). Candidaemia in Europe: Epidemiology and resistance. International Journal of Antimicrobial Agents, 27(5), 359–366. https://doi.org/10.1016/j.ijantimicag.2006.01.002
  • Trott, O., & Olson, A. J. (2010). AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. Journal of Computational Chemistry, 31(2), 455–461. https://doi.org/10.1002/jcc.21334
  • Tuccinardi, T. (2021). What is the current value of MM/PBSA and MM/GBSA methods in drug discovery? Expert Opinion on Drug Discovery, 16(11), 1233–1237. https://doi.org/10.1080/17460441.2021.1942836
  • Turner, P. (2005). XMGRACE, Version 5.1. 19. Center for Coastal and Land-Margin Research, Oregon Graduate Institute of Science and Technology, Beaverton.
  • Volkamer, A., Kuhn, D., Grombacher, T., Rippmann, F., & Rarey, M. (2012). Combining global and local measures for structure-based druggability predictions. Journal of Chemical Information and Modeling, 52(2), 360–372. https://doi.org/10.1021/ci200454v
  • Wager, T. T., Hou, X., Verhoest, P. R., & Villalobos, A. (2010). Moving beyond rules: The development of a central nervous system multiparameter optimization (CNS MPO) approach to enable alignment of druglike properties. ACS Chemical Neuroscience, 1(6), 435–449. https://doi.org/10.1021/cn100008c
  • Wager, T. T., Hou, X., Verhoest, P. R., & Villalobos, A. (2016). Central nervous system multiparameter optimization desirability: Application in drug discovery. ACS Chemical Neuroscience, 7(6), 767–775. https://doi.org/10.1021/acschemneuro.6b00029
  • Wang, C., Greene, D., Xiao, L., Qi, R., & Luo, R. (2017). Recent developments and applications of the MMPBSA method. Frontiers in Molecular Biosciences, 4(7), 87–18. https://doi.org/10.3389/fmolb.2017.00087
  • Wang, E., Sun, H., Wang, J., Wang, Z., Liu, H., Zhang, J. Z. H., & Hou, T. (2019). End-point binding free energy calculation with MM/PBSA and MM/GBSA: Strategies and applications in drug design. Chemical Reviews, 119(16), 9478–9508. https://doi.org/10.1021/acs.chemrev.9b00055
  • Wang, R., Lu, Y., & Wang, S. (2003). Comparative evaluation of 11 scoring functions for molecular docking. Journal of Medicinal Chemistry, 46(12), 2287–2303. https://doi.org/10.1021/jm0203783
  • Williams, C. J., Headd, J. J., Moriarty, N. W., Prisant, M. G., Videau, L. L., Deis, L. N., Verma, V., Keedy, D. A., Hintze, B. J., Chen, V. B., Jain, S., Lewis, S. M., Arendall, W. B., Snoeyink, J., Adams, P. D., Lovell, S. C., Richardson, J. S., & Richardson, D. C. (2018). MolProbity: More and better reference data for improved all-atom structure validation. Protein Science, 27(1), 293–315. https://doi.org/10.1002/pro.3330
  • Wisintainer, G. G. N. S., Simões, E., R., B., Lemos, T. L. G., Moura, S., Souza, L. G. S., Fonseca, A. M., Moraes, M. O., Pessoa, C., Roesch-Ely, M., & Henriques, J. A. P. (2014). Biflorin: An o-naphthoquinone of clinical significance. Anais Da Academia Brasileira De Ciencias, 86(4), 1907–1914. https://doi.org/10.1590/0001-3765201420140085
  • Wu, Z., Lei, T., Shen, C., Wang, Z., Cao, D., & Hou, T. (2019). ADMET evaluation in drug discovery. 19. Reliable prediction of human cytochrome P450 inhibition using artificial intelligence approaches. Journal of Chemical Information and Modeling, 59(11), 4587–4601. https://doi.org/10.1021/acs.jcim.9b00801
  • Yu, K., Geng, X., Chen, M., Zhang, J., Wang, B., Ilic, K., & Tong, W. (2014). High daily dose and being a substrate of cytochrome P450 enzymes are two important predictors of drug-induced liver injury. Drug Metabolism and Disposition: The Biological Fate of Chemicals, 42(4), 744–750. https://doi.org/10.1124/dmd.113.056267
  • Yuriev, E., Holien, J., & Ramsland, P. A. (2015). Improvements, trends, and new ideas in molecular docking: 2012–2013 in review. Journal of Molecular Recognition, 28(10), 581–604. https://doi.org/10.1002/jmr.2471
  • Zheng, M., Luo, X., Shen, Q., Wang, Y., Du, Y., Zhu, W., & Jiang, H. (2009). Site of metabolism prediction for six biotransformations mediated by cytochromes P450. Bioinformatics (Oxford, England), 25(10), 1251–1258. https://doi.org/10.1093/bioinformatics/btp140

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.