In Silico Antiprotozoal Evaluation of 1,4-Naphthoquinone Derivatives against Chagas and Leishmaniasis Diseases Using QSAR, Molecular Docking, and ADME Approaches
Abstract
:1. Introduction
2. Results and Discussion
2.1. QSAR Modeling
2.1.1. QSAR Model for Anti-chagas Activity
2.1.2. QSAR Model for Anti-L. amazonensis Activity
2.1.3. QSAR Model for anti-L. infantum Activity
2.1.4. QSAR Model for Toxicity
2.1.5. Validation of QSAR Models
2.1.6. Molecular Design and Applicability of QSAR Models
2.2. Molecular Docking
2.2.1. Docking in Trypanothione Reductase and Lanosterol α-Demethylase Proteins of T. Cruzi
2.2.2. Docking in Trypanothione Reductase, Arginase, and Aminotransferase Proteins of Leismania Genus
2.3. ADME Analysis
3. Materials and Methods
3.1. QSAR Modelling
3.1.1. In Vitro Anti-Chagas and Anti-Leishmaniasis Data
3.1.2. Development of Antiprotozoal QSAR Models
3.1.3. Validation of the QSAR Models
3.2. Molecular Docking
3.3. ADME Analysis
4. Conclusions
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
Appendix A
References
- Enfermedad de Chagas—OPS/OMS. Available online: https://www.paho.org/es/temas/enfermedad-chagas (accessed on 5 April 2022).
- Dos Santos Naujorks, A.A.; Da Silva, A.O.; Da Silva Lopez, R.; De Albuquerque, S.; Beatriz, A.; Marquez, M.R.; Pires De Lima, D. Novel naphthoquinone derivatives and evaluation of trypanocidal and leishmanicidal activities. J. Mater. Chem. B 2018, 6, 1–3. [Google Scholar] [CrossRef]
- Alonso-Padilla, J.; Rodríguez, A. High Throughput Screening for Anti–Trypanosoma cruzi Drug Discovery. PLoS Negl. Trop. Dis. 2014, 8, e3259. [Google Scholar] [CrossRef] [PubMed]
- Coura, J.R. Present situation and new strategies for Chagas disease chemotherapy: A proposal. Mem. Inst. Oswaldo Cruz 2009, 104, 549–554. [Google Scholar] [CrossRef] [Green Version]
- Schmunis, G.A.; Yadon, Z.E. Chagas disease: A new worldwide challenge. Acta Trop. 2010, 115, 14–21. [Google Scholar] [CrossRef] [PubMed]
- Arias, D.G.; Garay, A.S.; Rodrigues, D.; Forastieri, P.S.; Luna, L.E.; Bürgui, M.D.L.M.; Prieta, C.; Iglesia, A.A.; Cravero, R.M.; Guerrero, S.A. Rational design of nitrofuran derivatives: Synthesis and valuation as inhibitors of Trypanosoma cruzi trypanothione reductase. Eur. J. Med. Chem. 2017, 125, 1088–1097. [Google Scholar] [CrossRef] [PubMed]
- Urbina, J.A. Ergosterol biosynthesis and drug development for Chagas disease. Mem. Inst. Oswaldo Cruz 2009, 104, 311–318. [Google Scholar] [CrossRef] [Green Version]
- Valdez, R.H.; Düsman Tonin, L.T.; Ueda-Nakamura, T.; Dias Filho, B.P.; Monrgado-Diaz, J.A.; Sarragitto, M.H.; Vataru Nakamura, C. Biological activity of 1,2,3,4-tetrahydro-β-carboline-3-carboxamides against Trypanosoma cruzi. Acta Trop. 2009, 110, 7–14. [Google Scholar] [CrossRef]
- De Souza, E.M.; Lansiaux, A.; Bailly, C.; Wilson, W.D.; Hu, Q.; Boykin, D.W.; Batista, M.M.; Araújo-Jorge, T.C.; Soerio, M.N.C. Phenyl substitution of furamidine markedly potentiates its anti-parasitic activity against Trypanosoma cruzi and Leishmania amazonensis. Biochem. Pharmacol. 2004, 68, 593–600. [Google Scholar] [CrossRef]
- Rolim Neto, P.J.; Luiz Gomes, C.; Wanderley Sales, V.D.A.; de Melo, C.G.; Ferreira da Silva, R.M.; Vicente Nishimura, R.H.; Araújo, R. Beta-lapachone: Natural occurrence, physicochemical properties, biological activities, toxicity and synthesis. Phytochemistry 2021, 186, 112713. [Google Scholar] [CrossRef]
- Menna-Barreto, R.F.S.; Henriques-Pons, A.; Pinto, A.V.; Morgado-Diaz, J.A.; Soares, M.J.; De Castro, S.L. Effect of a β-lapachone-derived naphthoimidazole on Trypanosoma cruzi: Identification of target organelles. J. Antimicrob. Chemother. 2005, 56, 1034–1041. [Google Scholar] [CrossRef]
- Menna-Barreto, R.F.S.; Corrêa, J.R.; Pinto, A.V.; Soares, M.J.; De Castro, S.L. Mitochondrial disruption and DNA fragmentation in Trypanosoma cruzi induced by naphthoimidazoles synthesized from β-lapachone. Parasitol. Res. 2007, 101, 895–905. [Google Scholar] [CrossRef] [PubMed]
- Menna-Barreto, R.F.S.; Correa, J.R.; Cascabulho, C.M.; Fernandes, M.C.; Pinto, A.V.; Soares, M.J.; De Castro, S.L. Naphthoimidazoles promote different death phenotypes in Trypanosoma cruzi. Parasitology 2009, 136, 499–510. [Google Scholar] [CrossRef] [PubMed]
- Silva, L.R.; Souza Guimaraes, A.; Do Nascimento, J.; Do Santo Nascimento, I.J.; Barbosa da Silva, E.; McKerrow, J.H.; Cardoso, S.H.; da Silva-Júnior, E.F. Computer-aided design of 1,4-naphthoquinone-based inhibitors targeting cruzain and rhodesain cysteine proteases. Bioorg. Med. Chem. 2021, 41, 116213. [Google Scholar] [CrossRef] [PubMed]
- Ramos, E.I.; Garza, K.M.; Krauth-Siegel, R.L.; Bader, J.; Martinez, L.E.; Maldonado, R.A. 2,3-Diphenyl-1,4-Naphthoquinone: A potential chemotherapeutic agent against trypanosoma cruzi. J. Parasitol. 2009, 95, 461–466. [Google Scholar] [CrossRef] [Green Version]
- Becerra, N.A.; Espinosa-Bustos, C.; Vázquez, K.; Rivera, G.; Paulino, M.; Cantero, J.; Nogueda, B.; Chacón-Vargas, F.; Castillo-Velazquez, U.; Rodríguez, A.F.E.; et al. Expanding the chemical space of aryloxy-naphthoquinones as potential anti-Chagasic agents: Synthesis and trypanosomicidal activity. Med. Chem. Res. 2021, 30, 2256–2265. [Google Scholar] [CrossRef]
- Da Cunha-Júnior, E.F.; Pacienza-Lima, W.; Alves Ribeiro, G.; Daher Netto, C.; Do Canto-Cavalheiro, M.M.; Monteiro da Silva, A.J.; Ribeiro Costa, P.R.; Rossi-Bergamann, B.; Torres-Santo, E.C. Effectiveness of the local or oral delivery of the novel naphthopterocarpanquinone LQB-118 against cutaneous leishmaniasis. J. Antimicrob. Chemother. 2011, 66, 1555–1559. [Google Scholar] [CrossRef] [Green Version]
- De Araújo, M.V.; De Souza, P.S.O.; De Queiroz, A.C.; Da Matta, B.; Brandao Leite, A.; Da Silva, A.E.; de Franca, J.A.A.; Silva, T.M.S.; Camara, C.A.; Alexandre-Moreira, M.S. Synthesis, leishmanicidal activity and theoretical evaluations of a series of substituted bis-2-Hydroxy-1,4-Naphthoquinones. Molecules 2014, 19, 15180–15195. [Google Scholar] [CrossRef]
- Guimarães, T.T.; Pinto, M.C.F.R.; Lanza, J.S.; Melo, M.N.; Do Monte-Neto, R.L.; De Melo, I.M.M.; Diogo, E.B.T.; Ferreira, V.F.; Camara, C.A.; Valenca, W.O.; et al. Potent naphthoquinones against antimony-sensitive and -resistant Leishmania parasites: Synthesis of novel α- And nor-α-lapachone-based 1,2,3-triazoles by copper-catalyzed azide-alkyne cycloaddition. Eur. J. Med. Chem. 2013, 63, 523–530. [Google Scholar] [CrossRef]
- Teixeira, M.J.; De Almeida, Y.M.; Viana, J.R.; Holanda Filha, J.G.; Rodrigues, T.P.; Prata, J.R., Jr.; Coelho, I.C.B.; Rao, V.S.; Pompeu, M.M.L. In vitro and in vivo leishmanicidal activity of 2-hydroxy-3-(3-methyl-2-butenyl)-1,4-naphthoquinone (lapachol). Phytother. Res. 2001, 15, 44–48. [Google Scholar] [CrossRef]
- Lima, N.M.F.; Corraia, C.S.; Leon, L.L.; Machado, G.M.C.; De Fátima Madeir, A.M.; Santana, G.A.E.; Goulart, M.O.F. Antileishmanial activity of lapachol analogues. Mem. Inst. Oswaldo Cruz 2004, 99, 757–761. [Google Scholar] [CrossRef] [Green Version]
- Pandey, R.K.; Kumbhar, B.V.; Sundar, S.; Kunwar, A.; Prajapati, V.K. Structure-based virtual screening, molecular docking, ADMET and molecular simulations to develop benzoxaborole analogs as potential inhibitor against Leishmania donovani trypanothione reductase. J. Recept. Signal Transduct. 2017, 37, 60–70. [Google Scholar] [CrossRef] [PubMed]
- Kim, Y.S.; Park, S.Y.; Lee, H.J.; Suh, M.E.; Schollmeyer, D.; Lee, C.O. Synthesis and cytotoxicity of 6,11-Dihydro-pyrido- and 6,11-Dihydro-benzo [2,3-b]phenazine-6,11-dione derivatives. Bioorg. Med. Chem. 2003, 11, 1709–1714. [Google Scholar] [CrossRef]
- Da Silva Júnior, E.N.; Diogo, E.B.T.; Dias, G.G.; Rodrigues, B.L.; Guimarães, T.T.; Valenca, W.O.; Camara, C.A.; De Oliveira, M.R.N.; Camara, C.A.; De Oliveira, R.A.; et al. Synthesis and anti-Trypanosoma cruzi activity of Naphthoquinone-containing Triazoles: Electrochemical studies on the effects of the quinoidal moiety. Bioorg. Med. Chem. 2013, 21, 6337–6348. [Google Scholar] [CrossRef] [Green Version]
- Bahia, S.B.B.B.; Reis, W.J.; Jardim, G.A.M.; Souto, F.T.; De Simone, C.A.; Gatto, C.C.; Menna-Barreto, R.F.S.; De Castro, S.L.; Cavalvanti, B.C.; Pessoa, C.; et al. Molecular hybridization as a powerful tool towards multitarget quinoidal systems: Synthesis, trypanocidal and antitumor activities of naphthoquinone-based 5-iodo-1,4-disubstituted-, 1,4- and 1,5-disubstituted-1,2,3-triazoles. Med. Chem. Comm. 2016, 7, 1555–1563. [Google Scholar] [CrossRef]
- Pinto, A.V.; Pinto, C.N.; Pinto, M.D.C.F.R.; Rita, R.S.; Pezzella, C.A.C. Trypanocidal Activity of Synthetic Heterocyclic Derivatives of Active Quinones from Tabebuia sp. Arzneimittelforschung 1997, 47, 21045. [Google Scholar] [CrossRef]
- Jardim, G.A.M.; Silva, T.L.; Goulart, M.O.F.; De Simone, C.A.; Barbosa, J.M.C.; Salomão, K.; De Castro, S.L.; Bower, J.F.; Da Silva Júnio, R.E.N. Rhodium-catalyzed C-H bond activation for the synthesis of quinonoid compounds: Significant Anti-Trypanosoma cruzi activities and electrochemical studies of functionalized quinones. Eur. J. Med. Chem. 2017, 136, 406–419. [Google Scholar] [CrossRef] [Green Version]
- Ferreira, S.B.; Salomão, K.; De Carvalho da Silva, F.; Ventura Pinto, A.; Kaiser, C.R.; Pinto, A.C.; Ferreira, V.F.; De Castro, S.L. Synthesis and anti-Trypanosoma cruzi activity of β-lapachone analogues. Eur. J. Med. Chem. 2011, 46, 3071–3077. [Google Scholar] [CrossRef]
- Da Silva Júnior, E.N.; De Mel, I.M.M.; Diogo, E.B.T.; Costa, V.A.; De Souza Felho, J.D.; Valencia, W.O.; Camara, C.A.; De Oliveira, R.N.; De Araujo, A.S.; Emery, F.S.; et al. On the search for potential anti-Trypanosoma cruzi drugs: Synthesis and biological evaluation of 2-hydroxy-3-methylamino and 1, 2, 3-triazolic naphthoquinoidal compounds obtained by click chemistry reactions. Eur. J. Med. Chem. 2012, 52, 304–312. [Google Scholar] [CrossRef]
- Da Silva, E.N.; Menna-Barreto, R.F.S.; Pinto, M.C.F.R.; Silva, R.S.F.; Teixeira, D.V.; De Souza, C.B.V.; De Simone, C.A.; De Castro, S.L.; Ferreira, V.L.; Pinto, A.V. Naphthoquinoidal [1,2,3]-triazole, a new structural moiety active against Trypanosoma cruzi. Eur. J. Med. Chem. 2008, 43, 1774–1780. [Google Scholar] [CrossRef]
- Da Silva Júnior, E.N.; De Souza, M.C.; Fernandes, M.C.; Menna-Barreto, R.F.S.; Pinto, M.C.F.R.; De Assis Lopes, F.; de Simone, C.A.; Andrade, C.K.; Pinto, A.V.; Ferreira, V.F.; et al. Synthesis and anti-Trypanosoma cruzi activity of derivatives from nor-lapachones and lapachones. Bioorg. Med. Chem. 2008, 16, 5030–5038. [Google Scholar] [CrossRef]
- Ryu, C.K.; Kim, D.H. The antimicrobial activities of some 1,4-naphthalenediones (III). Arch. Pharmacal Res. 1993, 16, 161–163. [Google Scholar] [CrossRef]
- Salomão, K.; De Santana, N.A.; Molina, M.T.; De Castro, S.L.; Menna-Barreto, R.F.S. Trypanosoma cruzi mitochondrial swelling and membrane potential collapse as primary evidence of the mode of action of naphthoquinone analogues. BMC Microbiol. 2013, 13, 196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Valderrama, J.A.; Benites, J.; Cortés, M.; Pessoa-Mahana, H.; Prina, E.; Fournet, A. Studies on quinones. Part 38: Synthesis and leishmanicidal activity of sesquiterpene 1,4-quinones. Bioorg. Med. Chem. 2003, 13, 4713–4718. [Google Scholar] [CrossRef] [PubMed]
- Valderrama, J.A.; Zamorano, C.; González, M.F.; Prina, E.; Fournet, A. Studies on quinones. Part 39: Synthesis and leishmanicidal activity of acylchloroquinones and hydroquinones. Bioorg. Med. Chem. 2005, 13, 4153–4159. [Google Scholar] [CrossRef]
- Da Silva, A.J.M.; Netto, C.D.; Pacienza-Lima, W.; Torres-Santo, E.C.; Rosii-Bergmann, B.; Maurel, S.; Valentin, A.; Costa, P.R.R. Antitumoral, Antileishmanial and Antimalarial Activity of Pentacyclic 1,4-Naphthoquinone Derivatives. J. Braz. Chem. Soc. 2009, 20, 176–182. [Google Scholar] [CrossRef]
- De Araújo, M.V.; David, C.C.; Netto, J.C.; De Oliveira, L.A.P.L.; Da Silva, K.C.J.; Dos Santos, J.M.; Da Silva, J.K.S.; De A Brandão, V.B.C.; Silva, T.M.S.; Camara, C.A.; et al. Evaluation on the leishmanicidal activity of 2-N,N′-dialkylamino-1,4-naphthoquinone derivatives. Exp. Parasitol. 2017, 176, 46–51. [Google Scholar] [CrossRef]
- Da Silva, P.R.; De Oliveira, J.F.; Da Silva, A.L.; Marques Queiroz, C.; de Lima, M.D.C.A.; Sampaio Feitosa, A.P.; Araújo Duarte, D.M.; Da Silva, A.C.; Brelaz de Castro, M.C.A.; Alves Pereira, V.R.; et al. Novel indol-3-yl-thiosemicarbazone derivatives: Obtaining, evaluation of in vitro leishmanicidal activity and ultrastructural studies. Chem.-Biol. Interact. 2020, 315, 108899. [Google Scholar] [CrossRef]
- Scarim, C.B.; Jornada, D.H.; Machado, M.G.M.; Ferreira, C.M.R.; Dos Santos, J.L.; Chung, M.C. Thiazole, thio and semicarbazone derivatives against tropical infective diseases: Chagas disease, human African trypanosomiasis (HAT), leishmaniasis, and malaria. Eur. J. Med. Chem. 2019, 162, 378–395. [Google Scholar] [CrossRef]
- Pinto, E.G.; Santo, I.O.; Schmidt, T.J.; Borborema, S.E.T.; Ferreira, V.F.; Rocha, D.R.; Templone, A.G. Potential of 2-hydroxy-3-phenylsulfanylmethyl-[1,4]-naphthoquinones against Leishmania (L.) infantum: Biological activity and structure-activity relationships. PLoS ONE 2014, 9, e105127. [Google Scholar] [CrossRef]
- Tapia, R.A.; Tapia, R.A.; Prieto, Y.; Pautet, M.; Domard, M.; Domard, M.; Sarciron, M.E.; Walchshofer, N.; Walchshofer, N.; Fillion, H. Synthesis and Antileishmanial Activity of Indoloquinones Containing a Fused Benzothiazole Ring. Eur. J. Org. Chem 2002, 2002, 4005–4010. [Google Scholar] [CrossRef]
- Di Giorgio, C.; Delmas, F.; Filloux, N.; Robin, M.; Seferian, L.; Azas, N.; Gasquet, M.; Costa, M.; Timon-David, P.M.; Galy, J.P. In vitro activities of 7-substituted 9-chloro and 9-amino-2-methoxyacridines and their bis- and tetra-acridine complexes against Leishmania infantum. Antimicrob. Agents Chemother. 2003, 47, 174–180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cunha Araújo, I.A.; De Paula, R.C.; Alves, C.L.; Ferraz Faria, K.; De Oliveira, M.M.; Gonçalves Mendes, G.; Ferreira Abdia Dias, E.M.; Ribeiro, R.R.; De Oliveira, A.B.; Da Silva, S.M. Efficacy of lapachol on treatment of cutaneous and visceral leishmaniasis. Exp. Parasitol. 2019, 199, 67–73. [Google Scholar] [CrossRef] [PubMed]
- Dos Santos Faiões, V.; Da Frota, L.C.R.; Cunha-Junior, E.F.; Barcellos, J.C.F.; Da Silva, T.; Netto, C.D.; Gonçalves Da Silva, S.A.; Da Silva, A.J.M.; Costa, P.R.R.; Torres-Santos, E.C. Second-generation pterocarpanquinones: Synthesis and antileishmanial activity. J. Venom. Anim. Toxins Incl. Trop. Dis. 2018, 24, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jentzsch, J.; Koto, W.S.; Al Nasr, I.S.; Schobert, R.; Ersfeld, K.; Biersack, B. New Antiparasitic Bis-Naphthoquinone Derivatives. Chem. Biodivers. 2020, 17, e1900597. [Google Scholar] [CrossRef] [Green Version]
- Ramírez-Macías, I.; Marín, C.; Es-Samti, H.; Fernandéz, A.; Guardia, J.J.; Zentar, H.; Agil, a.; Chamboun, R.; Alvarez-Manzaneda, E.; Sánchez-Moreno, M. Taiwaniaquinoid and abietane quinone derivatives with trypanocidal activity against T. cruzi and Leishmania spp. Parasitol. Int. 2012, 61, 405–413. [Google Scholar] [CrossRef]
- Da Cruz, E.H.G.; Hussene, C.M.B.; Diasa, G.G.; Diogo, E.B.T.; De Melo, I.M.M.; Rodrigues, B.L.; Da Silva Júnior, E.N.; Da Silva, M.G.; Valencia, W.O.; Camara, C.A. 1,2,3-Triazole-, arylamino- and thio-substituted 1,4-naphthoquinones: Potent antitumor activity, electrochemical aspects, and bioisosteric replacement of C-ring-modified lapachones. Bioorg. Med. Chem. 2014, 22, 1608–1619. [Google Scholar] [CrossRef] [Green Version]
- Ramirez, O.; Motta-Mena, L.B.; Cordova, A.; Garza, K.M. A small library of synthetic Di-substituted 1, 4-naphthoquinones induces ROS-mediated cell death in murine fibroblasts. PLoS ONE 2014, 9, e106828. [Google Scholar] [CrossRef]
- Liu, Z.; Zhang, Z.; Zhang, W.; Yan, D. 2-Substituted-1-(2-morpholinoethyl)-1H-naphtho [2,3-d]imidazole-4,9-diones: Design, synthesis and antiproliferative activity. Bioorg. Med. Chem. Lett. 2018, 28, 2454–2458. [Google Scholar] [CrossRef]
- Baiju, T.V.; Almeida, R.G.; Sivanandam, S.T.; De Simone, C.A.; Brito, L.M.; Cavalcanti, B.C.; Pessoa, C.; Namboothiti, I.N.N.; Da Silva Júnior, E.N. Quinonoid compounds via reactions of lawsone and 2-aminonaphthoquinone with α-bromonitroalkenes and nitroallylic acetates: Structural diversity by C-ring modification and cytotoxic evaluation against cancer cells. Eur. J. Med. Chem. 2018, 151, 686–704. [Google Scholar] [CrossRef]
- Da Silva Júnior, E.N.; De Souza, M.C.B.V.; Pinto, A.V.; Pinto, C.C.; Goulart, M.O.F.; Barros, F.W.A.; Pessoa, C.; Costa-Lotufo, L.V.; Montenegro, R.C.; de Moraes, M.O.; et al. Synthesis and potent antitumor activity of new arylamino derivatives of nor-β-lapachone and nor-α-lapachone. Bioorg. Med. Chem. 2007, 15, 7035–7041. [Google Scholar] [CrossRef]
- Hoffmann, R.; Minkin, V.I.; Carpenter, B.K. Ockham’s Razor and chemistry. Int. J. Philos. Chem. 1997, 3, 3–28. [Google Scholar] [CrossRef]
- Laboratoire de Chemoinformatique. Available online: http://infochim.u-strasbg.fr/spip.php?rubrique41 (accessed on 29 August 2021).
- Wei Yap, C. PaDEL-Descriptor: An Open Source Software to Calculate Molecular Descriptors and Fingerprints. J. Comput. Chem. 2010, 31, 671–690. [Google Scholar] [CrossRef]
- Durant, J.L.; Leland, B.A.; Henry, D.R.; Nourse, J.G. Reoptimization of MDL keys for use in drug discovery. J. Chem. Inf. Model. 2002, 42, 1273–1280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martiní, J.R.V.; Ponce, Y.M.; Jacas, C.R.G.; Mayorga, K.M. QuBiLS—MAS, open source multi—platform software for atom-and bond-based topological descriptors computations. J. Cheminform. 2017, 9, 35. [Google Scholar] [CrossRef] [PubMed]
- Ghose, A.K.; Crippen, G.M. Atomic Physicochemical Parameters for Three-Dimensional-Structure-Directed Quantitative Structure-Activity Relationships. 2. Modeling Dispersive and Hydrophobic Interactions. J. Chem. Inf. Comput. Sci. 1987, 27, 21–35. [Google Scholar] [CrossRef] [PubMed]
- Klekota, J.; Roth, F.P. Chemical substructures that enrich for biological activity. Bioinformatics 2008, 24, 2518–2525. [Google Scholar] [CrossRef]
- Martínez, M.J.; Ponzoni, I.; Díaz, M.F.; Vazquez, G.E.; Soto, A.J. Visual analytics in cheminformatics: User—supervised descriptor selection for QSAR methods. J. Cheminform. 2015, 7, 39. [Google Scholar] [CrossRef] [Green Version]
- Garro Martinez, J.C.; Andrada, M.F.; Vega-Hissi, E.G.; Garibotto, F.M.; Nogueras, M.; Rodríguez, R.; Cobo, J.; Enriz, R.D.; Estrada, M.R. Dihydrofolate reductase inhibitors: A quantitative structure—activity relationship study using 2D-QSAR and 3D-QSAR methods. Med. Chem. Res. 2016, 26, 247–261. [Google Scholar] [CrossRef]
- Insubria QSAR PaDEL-Descriptor Model for Prediction of Endocrine Disruptors Chemicals (EDC) Estrogen Receptor (ER)-Binding Affinity. Available online: http://padel.nus.edu.sg/software/padeldescriptor/index.html (accessed on 5 October 2021).
- Samuelson, J. Why Metronidazole Is Active against both Bacteria and Parasites. ASM 1999, 43, 1533–1541. [Google Scholar] [CrossRef] [Green Version]
- Cardoso-Silva, J.; Papadatos, G.; Papageorgiou, L.G.; Tsoka, S. Optimal Piecewise Linear Regression Algorithm for QSAR Modelling. Mol. Inform. 2019, 38, 1800028. [Google Scholar] [CrossRef]
- Thurston, B.A.; Ferguson, A.L. Machine learning and molecular design of self-assembling-conjugated oligopeptides. Mol. Simul. 2018, 44, 930–945. [Google Scholar] [CrossRef]
- Ibrahum, Z.Y.; Uzairu, A.; Shallangwa, G.; Abechi, S. Theoretical design of novel antimalarial agents against P. falciparum strain, Dd2 through the QSAR modeling of synthesized 2′-substituted triclosan derivatives. Heliyon 2020, 6, e05032. [Google Scholar] [CrossRef] [PubMed]
- García-Jacas, C.R.; Barigye, S.J.; Marrero-Ponce, Y.; Acevedo-Martínez, L.; Valdés-Martiní, J.R.; Contreras-Torres, E. QuBiLS-MIDAS: A Parallel Free-Software for Molecular Descriptors Computation Based on Multilinear Algebraic Maps. J. Comput. Chem. 2014, 35, 1395–1409. [Google Scholar] [CrossRef] [PubMed]
- Ghose, A.K.; Viswanadhan, V.N.; Wendoloski, J.J. Prediction of Hydrophobic (Lipophilic) Properties of Small Organic Molecules Using Fragmental Methods: An Analysis of ALOGP and CLOGP Methods. J. Phys. Chem. 1998, 5639, 3762–3772. [Google Scholar] [CrossRef]
- Lanez, T.; Ahmedi, R. AlogP calculation of octanol-water partition coefficient of ferrocene derivatives. Int. J. Pharm.Tech. Res. 2015, 8, 408–414. [Google Scholar] [CrossRef] [Green Version]
- Prasanna, S.; Doerksen, R.J. Topological Polar Surface Area: A Useful Descriptor in 2D-QSAR. Curr. Med. Chem. 2009, 16, 21–41. [Google Scholar] [CrossRef]
- Organisation de Coopération et de Développement Economiques. Guidance Document on the Validation of (Quantitative)Structure-Activity Relationships [(Q)SAR] Models; OECD: Paris, France, 2007. [Google Scholar]
- Ciubotariu, D.; Medeleanu, M.; Vlaia, V.; Olariu, T.; Ciubotariu, C.; Dragos, D.; Corina, S. Molecular van der Waals Space and Topological Indices from the Distance Matrix. Molecules 2004, 9, 1053–1078. [Google Scholar] [CrossRef] [Green Version]
- Andrew, G.; Pablo, R. Chemometrics Applocations and Research QSAR in Medicinal Chemistry, 1st ed.; Appe Academic Press: Burlington, ON, Canada; Cambridge, MA, USA, 2016; pp. 366–381. [Google Scholar]
- Fernandes, J.; Gattass, C.R. Topological polar surface area defines substrate transport by multidrug resistance associated protein 1 (MRP1/ABCC1). J. Med. Chem 2009, 52, 1214–1218. [Google Scholar] [CrossRef]
- Gozalbes, R.; Doucet, J.P.; Derouin, F. Application of Topological Descriptors in QSAR and Drug Design: History and New Trends. Curr. Drug Targets Infect. Disord. 2002, 2, 93–102. [Google Scholar] [CrossRef]
- Tandon, H.; Ranjan, P.; Chakraborty, T.; Suhag, V. Polarizability: A promising descriptor to study chemical–biological interactions. Mol. Divers. 2021, 25, 249–262. [Google Scholar] [CrossRef]
- Putz, M.V.; Ionaşcu, C.; Putz, A.M.; Ostafe, V. Alert-QSAR. Implications for electrophilic theory of chemical carcinogenesis. Int. J. Mol. Sci. 2011, 12, 5098–5134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bhal, S.K.; Kassam, K.; Peirson, I.G.; Pearl, G.M. The rule of five revisited: Applying log D in place of log P in drug-likeness filters. Mol. Pharm. 2007, 4, 556–560. [Google Scholar] [CrossRef] [PubMed]
- Golbraikh, A.; Tropsha, A. Beware of q 2! J. Mol. Graph. Model. 2002, 20, 269–276. [Google Scholar] [CrossRef]
- Nossa, D.L.; Rozo Núñez, W.E.; Gómez Castaño, J.A.; Duchowicz, P.R. Antiprotozoal QSAR modelling for trypanosomiasis (Chagas disease) based on thiosemicarbazone and thiazole derivatives. J. Mol. Graph. Model. 2021, 103, 107821. [Google Scholar] [CrossRef] [PubMed]
- Roy, K. On some aspects of validation of predictive quantitative structure—Activity relationship models. Drug Discov. 2007, 2, 1567–1578. [Google Scholar] [CrossRef] [PubMed]
- López López, L.I.; Nery Flores, S.D.; Silva Belmares, S.Y.; Sáenz Galindo, A. Naphthoquinones: Biological properties and synthesis of lawsone and derivatives—A structured review. Vitae 2014, 21, 248–258. [Google Scholar]
- Rodrigues Coura, J.; De Castro, S.L. A critical review on chagas disease chemotherapy. Mem. Inst. Oswaldo Cruz 2002, 97, 3–24. [Google Scholar] [CrossRef]
- Silva, R.S.F.; De Amorim, M.B.C.; Pinto, F.R.; Emery, F.S.; Goulart, M.O.F.; Pinto, A.V. Chemoselective Oxidation of Benzophenazines by m-CPBA: N-Oxidation vs. Oxidative Cleavage. J. Braz. Chem. Soc. 2007, 18, 759–764. [Google Scholar] [CrossRef] [Green Version]
- Pacheco, P.A.F.; Galvão, R.M.S.; Faria, A.F.M.; Von Ranke, N.I.; Rangel, M.S.; Ribeiro, T.M.; Bello, M.I.; Rodrigues, C.R.; Ferreira, V.F.; Da Rocha, D.R.; et al. 8-Hydroxy-2-(1H-1,2,3-triazol-1-yl)-1,4-naphtoquinone derivatives inhibited P2X7 Receptor-Induced dye uptake into murine Macrophages. Bioorg. Med. Chem. 2019, 27, 1449–1455. [Google Scholar] [CrossRef]
- Chacón-Vargas, K.F.; Nogueda-Torres, B.; Sánchez-Torres, L.E.; Suarez-Contreras, E.; Villalobos-Rocha, J.C.; Torres-Martinez, Y.; Lara-Ramirez, E.E.; Fiorani, G.; Krauth-Siegel, R.L.; Bolognesi, M.L.; et al. Trypanocidal activity of quinoxaline 1,4 Di-N-oxide derivatives as trypanothione reductase inhibitors. Molecules 2017, 22, 220. [Google Scholar] [CrossRef] [Green Version]
- Bond, C.S.; Zhang, Y.; Berriman, M.; Cunningham, M.L.; Fairlamb, A.H.; Hunter, W.N. Crystal structure of Trypanosoma cruzi trypanothione reductase in complex with trypanothione, and the structure-based discovery of new natural product inhibitors. Structure 1999, 7, 81–89. [Google Scholar] [CrossRef] [Green Version]
- Chen, C.K.; Leung, S.F.; Guilbert, C.; Jacobson, M.P.; Mckerrow, J.H.; Podust, L.M. Structural characterization of CYP51 from Trypanosoma cruzi and Trypanosoma brucei bound to the antifungal drugs posaconazole and fluconazole. PLoS Negl. Trop. Dis. 2010, 4, e651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cardoso, M.F.C.; Forezi, L.S.M.; De Souza, A.S.; Faria, A.F.M.; Galvão, R.M.S.; Bello, M.L.; Da Silva, F.C.; Faria, R.X.; Ferreira, V.F. Tandem Synthesis of Furanaphthoquinones via Enamines and Evaluation of Their Antiparasitic Effects against Trypanosoma cruzi. J. Braz. Chem. Soc. 2022, 33, 238–250. [Google Scholar] [CrossRef]
- Inacio Filho, J.D. Estudo do Efeito da EGCG In Vitro e In Vivo, Suas Associações e Mecanismo Molecular de Ação em Leishmania Infantum. Ph.D. Thesis, Fundação Oswaldo Cruz, Rios de Janeiro, Brazil, 2018. [Google Scholar]
- Reynolds, K.A. Design and Synthesis of Quinoline, Cinchona Alkaloids and Other Potential Inhibitors or Leishmaniasis. Ph.D. Thesis, Griffith University, Brisbane, Australia, 2012. [Google Scholar]
- Braga, S.S. Multi-target drugs active against leishmaniasis: A paradigm of drug repurposing. Eur. J. Med. Chem. 2019, 183, 111660. [Google Scholar] [CrossRef] [PubMed]
- Ilari, A.; Baiocco, P.; Messori, L.; Fiorillo, A.; Boffi, A.; Gramiccia, M.; Di Muccio, T.; Colotti, G. A gold-containing drug against parasitic polyamine metabolism:The X-ray structure of trypanothione reductase from Leishmania infantum in complex with auranofin reveals a dual mechanism of enzyme inhibition. Amino Acids 2012, 42, 803–811. [Google Scholar] [CrossRef] [Green Version]
- Venkatesan, S.K.; Shukla, A.K.; Dubey, V.K. Molecular Docking Studies of Selected Tricyclic and Quinone Derivatives on Trypanothione Reductase of Leishmania infantum. J. Comp. Chem. 2010, 32, 174–182. [Google Scholar] [CrossRef]
- Manjolin, L.C.; Dos Reis, M.B.G.; Do Carmo Maquiaveli, C.; Santos-Filho, O.A.; Da Silva, E.R. Dietary flavonoids fisetin, luteolin and their derived compounds inhibit arginase, a central enzyme in Leishmania (Leishmania) amazonensis infection. Food Chem. 2013, 141, 2253–2262. [Google Scholar] [CrossRef] [Green Version]
- De Lima, E.C.; Castelo-Branco, F.S.; Maquiaveli, C.C.; Farias, A.B.; Rennó, M.N.; Boechat, N.; Silva, E.R. Phenylhydrazides as inhibitors of Leishmania amazonensis arginase and antileishmanial activity. Bioorg. Med. Chem. 2019, 27, 3853–3859. [Google Scholar] [CrossRef]
- Kanyo, Z.F.; Scolnick, L.R.; Ash, D.E.; Christianson, D.W. Structure of unique binuclear manganese cluster in arginase. Lett. Nat. 1996, 383, 554–557. [Google Scholar] [CrossRef]
- Estrada, E.; Molina, E. Novel local (fragment-based) topological molecular descriptors for QSPR / QSAR and molecular design. J. Mol. Graph. Model. 2001, 20, 54–64. [Google Scholar] [CrossRef]
- Daina, A.; Michielin, O.; Zoete, V. SwissADME: A free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci. Rep. 2017, 7, 42717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zafar, F.; Gupta, A.; Thangavel, K.; Khayana, K.; Sani, A.A.; Ghosal, A.; Tandon, P.; Nishat, N. Physicochemical and Pharmacokinetic Analysis of Anacardic Acid Derivatives. ACS Omega 2020, 5, 6021–6030. [Google Scholar] [CrossRef] [PubMed]
- Bhal, S.K. Advanced Chemistry Development (ACD/LABS). Log P—Making Sense of the Value; Advanced Chemistry Development: Toronto, ON, Canada, 2007. [Google Scholar]
- Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feeney, P.J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Deliv. Rev. 2012, 64, 4–17. [Google Scholar] [CrossRef]
- Mahanthesh, M.T.; Ranjith, D.; Raghavendra, Y.; Jyothi, R.; Narappa, G.; Ravi, M.V. Swiss ADME prediction of phytochemicals present in Butea monosperma (Lam.) Taub. J. Pharmacogn. Phytochem. 2020, 9, 1799–1809. [Google Scholar]
- Delaney, J.S. ESOL: Estimating aqueous solubility directly from molecular structure. J. Chem. Inf. Comput. Sci. 2004, 44, 1000–1005. [Google Scholar] [CrossRef]
- Ali, P.; Camilleri, P.; Brown, M.B.; Hutt, A.J.; Kirton, S.B. Revisiting the general solubility equation: In silico prediction of aqueous solubility incorporating the effect of topographical polar surface area. J. Chem. Inf. Model. 2012, 52, 420–428. [Google Scholar] [CrossRef]
- Löbenberg, R.; Amidon, G.L.; Ferraz, H.G.; Bou-Chacra, N. Mechanism of gastrointestinal drug absorption and application in therapeutic drug delivery. In Therapeutic Delivery Methods: A Concise Overview of Emerging Areas; Future Science Ltd.: London, UK, 2013; pp. 8–22. [Google Scholar] [CrossRef]
- Osorio, E.J.; Robledo, S.M.; Arango, G.J.; Muskus, C.E. Leishmania: Papel de la glicoproteína P en la mediación de resistencia a medicamentos y estrategias de reversión. Biomédica 2005, 25, 242. [Google Scholar] [CrossRef] [Green Version]
- Finch, A.; Pillans, P. P-glycoprotein and its role in drug-drug interactions. Aust. Prescr. 2014, 37, 137–139. [Google Scholar] [CrossRef]
- Daina, A.; Zoete, V. A BOILED-Egg To Predict Gastrointestinal Absorption and Brain Penetration of Small Molecules. Chem. Pub. Soc. Eur. 2016, 11, 1117–1121. [Google Scholar] [CrossRef] [Green Version]
- Potts, R.O.; Guy, R.H. Predicting Skin Permeability. Pharm. Res. 1992, 9, 663–669. [Google Scholar] [CrossRef]
- Ranjith, D.; Ravikumar, C. SwissADME predictions of pharmacokinetics and drug-likeness properties of small molecules present in Ipomoea mauritiana Jacq. J. Pharmacogn. Phytochem. 2019, 8, 2063–2073. Available online: http://www.swissadme.ch/index.php (accessed on 30 April 2022).
- Jaimes-Santoyo, J.; De Montesinos-Sampedro, A.; Barbosa-Cobos, R.E.; Moreno-Mutio, S.G.; Rodriguez-Ballesteros, D.; Ramos-Cervantes, T.; Ocharán-Hernández, M.E.; Toscano-Garibay, J.; Beltrán-Ramírez, O. El Citocromo P-450. Rev. Hosp. Juárez Mex. 2014, 81, 250–256. [Google Scholar]
- Deodhar, M.; Al Rihani, S.B.; Arwood, M.J.; Drakjian, L.; Dow, P.; Turgeon, J.; Michaud, V. Mechanisms of cyp450 inhibition: Understanding drug-drug interactions due to mechanism-based inhibition in clinical practice. Pharmaceutics 2020, 12, 846. [Google Scholar] [CrossRef] [PubMed]
- Ogu, C.C.; Maxa, J.L. Drug Interactions Due to Cytochrome P450. Bayl. Univ. Med. Cent. 2017, 8280, 11–14. [Google Scholar] [CrossRef]
- Ghose, A.K.; Viswanadhan, V.N.; Wendoloski, J.J. A Knowledge-Based Approach in Designing Combinatorial or Medicinal Chemistry Libraries for Drug Discovery. 1. A Qualitative and Quantitative Characterization of Known Drug Databases. J. Comb. Chem. 1999, 1, 55–68. [Google Scholar] [CrossRef]
- Veber, D.F.; Johnson, S.R.; Cheng, H.J.; Smith, B.R.; Ward, K.W.; Kopple, K.D. Molecular Properties That Influence the Oral Bioavailability of Drug Candidates. J. Med. Chem. 2002, 45, 2615–2623. [Google Scholar] [CrossRef]
- Egan, W.J.; Merz, K.M.; Baldwin, J.J. Prediction of Drug Absorption Using Multivariate Statistics. J. Med. Chem. 2000, 43, 3867–3877. [Google Scholar] [CrossRef]
- Muegge, I.; Heald, S.L.; Brittelli, D. Simple Selection Criteria for Drug-like Chemical Matter. J. Med. Chem. 2001, 44, 1841–1846. [Google Scholar] [CrossRef]
- Martin, Y.C. A Bioavailability Score. J. Med. Chem. 2005, 48, 3164–3170. [Google Scholar] [CrossRef]
- ACD/ChemSketch; Versión 2021.1.3; Advanced Chemistry Development, Inc.: Toronto, ON, Canada, 2021; Available online: www.acdlabs.com (accessed on 30 April 2022).
- Mold2_FDA. Available online: https://www.fda.gov/science-research/bioinformatics-tools/mold2 (accessed on 29 August 2021).
- Laboratory of Chemoinformatique. 2013. Available online: http://infochim.u-strasbg.fr/ (accessed on 30 April 2022).
- Mercader, A.G.; Duchowicz, P.; Ferna, F. Replacement Method and Enhanced Replacement Method Versus the Genetic Algorithm Approach for the Selection of Molecular Descriptors in QSPR/QSAR Theories. J. Chem. Inf. Model. 2010, 50, 1542–1548. [Google Scholar] [CrossRef]
- Mathworks, MATLAB—Mathworks—MATLAB & Simulink. 2016. Available online: https://nl.mathworks.com/products/matlab.html%0Ahttp://www.mathworks.com/products/matlab/ (accessed on 29 August 2021).
- Gramatica, P. Principles of QSAR models validation: Internal and external. QSAR Comb. Sci. 2007, 26, 694–701. [Google Scholar] [CrossRef]
- Veerasamy, R.; Rajak, H.; Jain, A.; Sivadasan, S.; Varghese, C.P.; Agrawal, R.K. Validation of QSAR Models—Strategies and Importance. Int. J. Drug Des. Discov. 2011, 2, 511–519. [Google Scholar]
- Singh, B.K.; Sarkar, N.; Jagannadham, M.V.; Dubey, V.K. Modeled structure of trypanothione reductase of Leishmania infantum. BMB Rep. 2008, 41, 444–447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baiocco, P.; Colotti, G.; Franceschini, S.; Ilari, A. Molecular Basis of Antimony Treatment in Leishmaniasis. J. Med. Chem. 2009, 52, 2603–2612. [Google Scholar] [CrossRef]
- Antonio, E.L.D.; Ullman, B.; Roberts, S.C.; Dixit, U.P.; Wilson, M.E.; Hai, Y.; Christianson, D.W. Crystal structure of arginase from Leishmania mexicana and implications for the inhibition of polyamine biosynthesis in parasitic infections. Arch. Biochem. Biophys. 2013, 535, 163–176. [Google Scholar] [CrossRef] [Green Version]
- Moreno, M.A.; Abramov, A.; Abendroth, J.; Alonso, A.; Zhang, S.; Alcolea, P.J.; Edwards, T.; Lorimer, D.; Myley, P.J.; Larraga, V. Structure of tyrosine aminotransferase from Leishmania infantum. Acta Crystallogr. 2014, 70, 583–587. [Google Scholar] [CrossRef] [Green Version]
- Molexus, Molegro Virtual Docker-Manual, Copyrigh, M. 2019. Available online: http://molexus.io/molegro-virtual-docker/ (accessed on 30 April 2022).
- Filgueira De Azevedo, W. Docking Screens for Drug Discovery; Humana Press: Ria Grande Do Sul, Brazil, 2019. [Google Scholar]
- Frisch, J.P.M.J.; Trucks, G.W.; Schlegel, H.B.; Scuseria, G.E.; Robb, M.A.; Cheeseman, J.R.; Zakrzewski, V.G.; Montgomery, J.A., Jr.; Stratmann, R.E.; Burant, J.C.; et al. Gaussian 98, Revision A. 7; Gaussian Inc.: Pittsburgh, PA, USA, 1998; p. 49. [Google Scholar]
- Thomsen, R.; Christensen, M.H. MolDock: A New Technique for High-Accuracy Molecular Docking. J. Med. Chem. 2006, 49, 3315–3321. [Google Scholar] [CrossRef]
- De Azevedo, W.F., Jr. MolDock Applied to Structure-Based Virtual Screening. Curr. Drug Targets 2010, 11, 327–334. [Google Scholar] [CrossRef]
- Da Cunha, E.F.F.; Azevedo Martins, R.C.; Girão Albuquerque, M.; de Alencastro, R.B. LIV-3D-QSAR model for estrogen receptor ligands. J. Mol. Model. 2004, 10, 297–304. [Google Scholar] [CrossRef]
Descriptor | Name | Short Description | Type | |
---|---|---|---|---|
a1 | frag16 | Fingerprint | ||
a2 | MACCSFP72 | (Any bond; Red: Oxygen; Gray:Any atom) | Fingerprint | |
a3 | K_Q_AB_nCi_2_SS10_T_KA_r_MAS | Refractivity | 2D | |
a4 | K_B_AB_nCi_2_DS7_C_KA_e-p_MAS | Electronegativity/Polarizability | 2D | |
a5 | K_B_AB_nCi_2_DS2_X_KA_r-c_MAS | Refractivity/Charge | 2D | |
a6 | N2_F_AB_nCi_2_NS7_P_KA_m_MAS | Mass | 2D |
Descriptor | Name | Short Description | Type | |
---|---|---|---|---|
b1 | KRFP2 | (Klekota–Roth fingerprint, presence of chemical substructures) | Fingerprint | |
b2 | minHBint9 | Electro-topological state atom type descriptor (minimal strength E-state type descriptor for potential hydrogen bonds of path length 9) | 2D | |
b3 | IC3 | Information content index (3-order neighborhood symmetry) | 2D | |
b4 | MDEC-23 | Molecular edge distance | 2D |
Descriptor | Name | Short Description | Type |
---|---|---|---|
c1 | MATS3c | Moran Correlation—lag 3/load-weighted | 2D |
c2 | nHBint7 | Atom-like electro-topological state | 2D |
c3 | AM_F_AB_nCi_2_NS12_T_KA_a_MAS | Alog P (partition) | 2D |
c4 | AM_B_AB_nCi_2_NS2_C_KA_psa-v_MAS | C atoms in aliphatic chain Topological area of the polar surface/Vdw volume | 2D |
c5 | AM_B_AB_nCi_2_SS1_C_KA_v-c_MAS | Volume of Vdw/Charge | 2D |
c6 | AM_Q_AB_nCi_2_NS15_X_KA_a_MAS | Heteroatom–Partitioning Algorithm (Alog P) | 2D |
Descriptor | Name | Short Description | Type |
---|---|---|---|
d1 | K_B_AB_nCi_2_NS3_T_KA_psa-r_MAS | Topological polar surface area; refractivity | 2D |
d2 | K_B_AB_nCi_2_DS7_P_KA_c-p_MAS | Aromatic C atoms. Charge; polarization | 2D |
d3 | N2_B_AB_nCi_2_MP4_P_KA_psa-p_MAS | Aromatic C atoms. Topological polar surface area; polarization | 2D |
d4 | K_B_AB_nCi_2_DS3_X_KA_a-e_MAS | Heteroatoms. Partition algorithm (Log P); electronegativity | 2D |
d5 | K_Q_AB_nCi_2_SS14_C_KA_c_MAS | C atoms in aliphatic chain. Charge. | 2D |
Parameter | QSAR Models | ||||
---|---|---|---|---|---|
Anti-Chagas (Ec. 1) | anti-L. amazonensis (Ec. 2) | Anti-L. infantum (Ec. 3) | Toxicity (Ec. 4) | ||
98% | 100% | ||||
107 | 48 | 72 | 60 | 60 | |
23 | 6 | 9 | 8 | 8 | |
23 | 6 | 9 | 7 | 8 | |
0.83 | 0.78 | 0.66 | 0.91 | 0.70 | |
0.48 | 0.38 | 0.35 | 0.22 | 0.32 | |
0.40 | 0.19 | 0.79 | 0.44 | 0.44 | |
0.71 | 0.70 | 0.66 | 0.92 | 0.92 | |
0.54 | 0.35 | 0.46 | 0.18 | 0.18 | |
0.06 | 0.09 | 0.08 | 0.08 | 0.09 | |
0.10 | 0.59 | 0.74 | 0.74 | 0.95 | |
k | 1.12 | 1.03 | 1.03 | 0.96 | 0,97 |
k’ | 0.86 | 0.88 | 0.92 | 1.03 | 1.00 |
0.80 | 0.80 | 0.71 | 0.89 | 0.75 | |
0.76 | 0.77 | 0.65 | 0.82 | 0.63 | |
0.77 | 0.65 | 0.80 | 0.88 | 0.75 | |
CCC | 0.86 | 0.86 | 0.79 | 0.89 | 0.83 |
0 | 0 | 0 | 0 | 1 |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Prieto Cárdenas, L.S.; Arias Soler, K.A.; Nossa González, D.L.; Rozo Núñez, W.E.; Cárdenas-Chaparro, A.; Duchowicz, P.R.; Gómez Castaño, J.A. In Silico Antiprotozoal Evaluation of 1,4-Naphthoquinone Derivatives against Chagas and Leishmaniasis Diseases Using QSAR, Molecular Docking, and ADME Approaches. Pharmaceuticals 2022, 15, 687. https://doi.org/10.3390/ph15060687
Prieto Cárdenas LS, Arias Soler KA, Nossa González DL, Rozo Núñez WE, Cárdenas-Chaparro A, Duchowicz PR, Gómez Castaño JA. In Silico Antiprotozoal Evaluation of 1,4-Naphthoquinone Derivatives against Chagas and Leishmaniasis Diseases Using QSAR, Molecular Docking, and ADME Approaches. Pharmaceuticals. 2022; 15(6):687. https://doi.org/10.3390/ph15060687
Chicago/Turabian StylePrieto Cárdenas, Lina S., Karen A. Arias Soler, Diana L. Nossa González, Wilson E. Rozo Núñez, Agobardo Cárdenas-Chaparro, Pablo R. Duchowicz, and Jovanny A. Gómez Castaño. 2022. "In Silico Antiprotozoal Evaluation of 1,4-Naphthoquinone Derivatives against Chagas and Leishmaniasis Diseases Using QSAR, Molecular Docking, and ADME Approaches" Pharmaceuticals 15, no. 6: 687. https://doi.org/10.3390/ph15060687