The extent of drug action is in direct relationship with the amount of the drug in aqueous medium in contact with the substrate molecules. The factors affecting this concentration in a biological system can be classified into the pharmacokinetics (PK) phase and the pharmacodynamics phase of drug action. Thus, this research focused on the in silico PKs analyses of phenolics (furocoumarinic acid, liquiritin, isorhamnetin and syringin) identified from ethylacetate fraction of methanol leaf extract of A. conyzoides. Determination of the pharmacokinetics and physicochemical properties of the above phenolics were achieved using SwissADME, ADMETlab 2.0 and SuperCYPsPred webservers synergistically. The phenolics with best in-silico pharmacokinetics parameters was further studied experimentally using in-vitro α-amylase and α-glucosidase inhibition assays. The findings indicated that four phenolics are soluble in water, and all the ligands have consensus logP values less than 5 according to Lipinski’s Rule of 5, with isorhamnetin being the best (LogP 1.6). Also, most of the phenolics are non-inhibitors of the main CYP450 isozymes, but 1A2 and 3A4 were inhibited by isorhamnetin. Similarly, they are mostly non-substrates of the isozymes, but 2C9, 2C19, and 2D6 were metabolized by isorhamnetin. Inhibition assays using isorhamnetin-containing sample indicated that the inhibitory effects were more on α-glucosidase (IC50 of 18.11 and 15.97µg/ml for acarbose and isorhamnetin, respectively) than on α-amylase. This study has demonstrated that these phenolics from ethylacetate fraction of methanol leaf extract of A. conyzoides have relatively good pharmacokinetics within the acceptable limit of drug-like molecules.
pharmacokinetics, in-silico, Ageratum conyzoides, phenolics, ethylacetate
Adel, M.N., Asma, E., Moayed, B.M., Zhai, E.L., et al. (2023). “Rigidity and flexibility of pyrazole, s-triazole, and v-triazole derivative of chloroquine as potential therapeutic against COVID-19”. Journal of Medical and Chemical Science, 6(9), 2056-2084. https://doi.org/10.1155/2023/4562974.
Alam, S., & Khan, F. (2018). Virtual screening, docking, admet and system pharmacology studies on garcinia caged xanthone derivatives for anticancer activity. Scientific reports, 8, 5524.
Banerjee, P., Dunkel, M., Kemmler, E., & Preissner, R. (2020). SuperCYPsPred—a web server for the prediction of cytochrome activity. Nucleic Acids Research, 48(W1), W580- W585. https://doi.org/10.1093/nar/gkaa166.
Bidstrup, T.B., Bjornsdottir, I., Sidelmann, U.G., Thomsen, M.S., & Hansen, K. T. (2003). CYP2C8 and CYP3A4 are the principal enzymes involved in the human in-vitro biotransformation of the human insulin scretagogue, repaglinide. British Journal of Clinical Pharmacology, 56,305-314.
Chen, F., Li, D., Zhang, B., Sun, J., Sun, F., Ji, X., et al. (2018). Alterations of drug-metabolizing enzymes and transporters under diabetic conditions: what is the potential clinical significance? Drug Metabolism Reviews https://doi.org/10.1080/03602532.2018.1497645.
Chen, S.Z., Pan, P.P., Shen, L.B., Hu, G.X., et al. (2014). Drug-drug interaction of losartan and glimepiride metabolism by recombinant microsome CYP2C9*1, 2C9*3, 2C9*13, and 2C9*16 in vitro. International Journal of Clinical Pharmacology Therapeutics, 52, 732–738.
Daina, A., Olivier, M., & Vincent, Z. (2017). SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Scientific Report, 7, 42717.
Daio, X., Carlier, J., Zhu, M., Pang, S., Kronstrand, R., Scheidweiler, K.B., & Huestis, M.A. (2017). In vitro and in vivo human metabolism of a new synthetic cannabinoid NM-2201 (CBL-2201). Forensic Toxicology, 35, 20-32.
Di, L. (2014). The role of drug metabolizing enzymes in clearance. Expert Opinion in Drug Metabolism and. Toxicology, 10, 379-393.
Do, M.T., Kim, H.G., Tran, T.T., Khanal, T., Choi, J.H., Chung, Y.C., Jeong, T.C., & Jeong, H.G. (2014). Metformin suppresses CYP1A1 and CYP1B1 expression in breast cancer cells by down-regulating aryl hydrocarbon receptor expression. Toxicology Application in Pharmacolog, 280,138-148.
Ghani, U. (2015). Re-exploring promising š¼-glucosidase inhibitors for potential development into oral anti-diabetic drugs: finding needle in the haystack. Europian Journal of Medicinal Chemistry, 103, 133-162. https://doi.org/10.1016/j.ejmech.2015.08.043.
Guang-Zhi, D., Ju-Hee, L., Sung-Hwan, K., Ji-Hye, Y., Il-Je, C., Seung, H.K., et al. (2014). AMPK activation by isorhamnetin protects hepatocytes against oxidative stress and mitochondrial dysfunction. European Journal of Pharmacology, 740, 634-640. https://doi.org/10.1016/j.ejphar.2014.06.017
Hirota, T., Eguchi, S., & Ieiri, I. (2013). Impact of genetic polymorphisms in CYP2C9 and CYP2C19 on the pharmacokinetics of clinically used drugs. Drug Metabolism and Pharmacokinetic, 28, 28-37.
Huang, S.M. Strong, J.M., Zhang, L., Reynolds, K.S., Nallani, S., Temple, R., et al. (2008). New era in drug interaction evaluation: US Food and Drug Administration update on CYP enzymes, transporters, and the guidance process. Journal of Clinical Pharmacology, 48 (6), 662-670. https://doi.org/10.1177/0091270007312153.
Jin-Jing, Z., Jin-Qing, S., Shu-Yi, P., & Kai, W. (2016). Treatment with isorhamnetin protects the brain against ischemic injury in mice. Neurochemistry Research, 41(8),1939-1948.
Kato, H. (2019). Computational prediction of cytochrome P450 inhibition and induction. Drug Metabolism and Pharmacokinetic, 35(1):30-44. https://doi.org/10.1016/j.dmpk.2019.11.006.
Kazmi, S.R., Jun, R., Yu, M.S., Jung, C., & Na, D. (2019). In silico approaches and tools for the prediction of drug metabolism and fate: A review. Computers in Biology and Medicine, 106, 54-64. https://doi.org/10.1016/j.compbiomed.2019.01.008
Kim, S., Thiessen, P.A., Bolton, E.E., Chen, J., Fu, G., Gindulyte, A., Han, L., et al. (2016). PubChem Substance and Compound databases. Nucleic acids research, 44(D1), D1202-D1213.
Kim, Y.C., Lee, A.K., Lee, J.H., Lee, I., Lee, D.C., Kim, S.H., Kim, et al. (2005). Pharmacokinetics of theophylline in diabetes mellitus rats: induction of CYP1A2 and CYP2E1 on 1,3-dimethyluric acid formation. European Journal of Pharmaceutical Science, 26, 114-123.
Kirchmair, J. Goller, A.H., Lang, D., Kunze, J., Testa, B., Wilson, I.D., et al. (2015). Predicting drug metabolism: experiment and/or computation? Nature Review and Drug Discovery, 14, 387-404. https://doi.org/10.1038/nrd4581.
Kobori, T., Harada, S., Nakamoto, K., & Tokuyama, S. (2013). Functional alterations of intestinal P-glycoprotein under diabetic conditions. Biological and Pharmaceutical Bulletin, 36, 1381-1390.
Koto-te-Nyiwa, N., Masengo, C.A., & Pius, T.M. (2015). Ethno-botany and Pharmacognosy of Ageratum conyzoides L. (Compositae). Journal of Advancement in Medical and Life Sciences, 2(4), 1-6.
Lee, D.Y., Chung, H.J., Choi, Y.H., Lee, U., Kim, S.H., Lee, I., & Lee, M.G. (2009). Pharmacokinetics of ipriflavone and its two metabolites, M1 and M5, after the intravenous and oral administration of ipriflavone to rat model of diabetes mellitus induced by streptozotocin. European Journal of Pharmaceutical Science, 38, 465-471.
Li, Z.H., Deng, Y., Cai, H.L., Guo, Z.H., Hou, Z.Y., Wu, G., et al. (2016). Pharmacokinetic properties and bioequivalence of spironolactone tablets in fasting and fed healthy Chinese male subjects. International Journal of Clinical Pharmacology and Therapeutics, 54(6), 455–461.
Liang, Y., Li, S., & Chen, L. (2015). The physiological role of drug transporters. Protein Cell, 6, 334-350.
Lin, J.H., & Yamazaki, M. (2003). Role of P-glycoprotein in pharmacokinetics: clinical implications. Clinical Pharmacokinetic, 42, 59-98.
Lipinski, C.A., Lombardo, F., Dominy, B.W., & Feeney, P.J. (2001). Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Advance in Drug Delivery and Review, 46, 3-26.
Liqing, H., Huan, H., Zhantu, L., Dan, L., Dong, Y., & Ming, H. (2016). Protective effects of isorhamnetin on cardiomyocytes against anoxia/reoxygenation[1]induced injury is mediated by SIRT1. Journal of Cardiovascular Pharmacology, 67, 526-537.
Liu, L., & Liu, X.D. (2014). Alterations in function and expression of ABC transporters at blood-brain barrier under diabetes and the clinical significances. Frontier Pharmacology, 5, 273.
Marilena, A., Janet, A.G., Carlos, M., & Sergio, O.S.S. (2015). Topical anti-inflammatory effects of isorhamnetin glycosides isolated from Opuntia ficus-indica. Biomedical Research International, 30-41. 10.1155/847320.
Martignoni, M., Groothuis, G.M., & de Kanter, R. (2006). Species differences between mouse, rat, dog, monkey and human CYP-mediated drug metabolism, inhibition and induction. Expert Opinion in Drug Metabolism and Toxicology, 2, 875–894.
Matzke, G.R., Frye, R.F., Early, J.J., Straka, R.J., & Carson, S.W. (2000). Evaluation of the influence of diabetes mellitus on antipyrine metabolism and CYP1A2 and CYP2D6 activity. Pharmacotherapy, 20, 182–190.
Mitra, A., Tamil, I., Dineshkumar, B., Nandhakumar, M., & Senthilkumar, M. (2010). In vitro study on α-amylase inhibitory activity of an Indian medicinal plant, Phyllanthus amarus. Indian Journal of Pharmacy, 42, 280.
Ottaviani, G., Gosling, D. J., Patissier, C., Rodde, S., Zhou, L., & Faller, F. (2010). What is modulating solubility in simulated intestinal fluids? European Journal of Pharmaceutical Science, 41, 452-457. https://doi.org/10.1016/j.ejps.2010.07.012.
Ozioko, P.C., Ibrahim, A., Muhammad, Y.Y., & Bashir, M. (2024). Fractionation and Identification of Phenolic Compounds from Ethylacetate Fraction of Ageratum conyzoides Methanolic Leaf Extract. International Journal of Emerging Multidisciplinarie: Biomedical and Clinical Research, 2(1), 1-21 https://doi.org/10.54938/ijemdbmcr.2024.02.1.272
Ozioko, P.C., Muhammed, Y.Y., & Ibrahim, A. (2022). Ageratum conyzoides Methanol Leaf Extract: Phytochemicals with Antidiabetic Potential via Antioxidant Activity. Asian Journal of Biochemistry, 17(1), 15-24. https://doi.org/10.3923/ajb.2022.15.24
Palleria, C., Di Paolo, A., Giofrè, C., Caglioti, C., Leuzzi, G., Siniscalchi, A., et al. (2013). Pharmacokinetic drug-drug interaction and their implication in clinical management. Journal of research in medical sciences: the official journal of Isfahan University of Medical Sciences, 18(7), 601–610. PMID: 24516494
Pajouhesh, H., & Lenz, G.R. (2005). "Medicinal Chemical Properties of Successful Central Nervous System Drugs". NeuroRx., 2(4), 541-553. https://doi.org/10.1602/neurorx.2.4.541
Pistia-Brueggeman, G., & Hollingsworth, R.I. (2001). A preparation and screening strategy for glycosidase inhibitors. Tetrahedron, 57, 8773-8778.
Rahman, M.A., Akter, N., Rashid, H., Ahmed, N.U., & Islam, M.S. (2012). Analgesic and antiāinflammatory effect of whole Ageratum conyzoides and Emilia sonchifolia alcoholic extracts in animal models. African Journal of Pharmacy and Pharmacology, 6, 1469-1476.
Ritchie, T.J., Macdonald, S.J.F., Peace, S., Pickett, S.D., & Luscombe, C.N. (2013). Increasing small molecule drug developability in suboptimal chemical space. Medicinal Chemistry Communication, 4, 673. 673-680
Shaaban, S., Abdou, A., Alhamzani, A.G., Abou-Krisha, M.M., Al-Qudah, M.A., Alaasar, M., et al. (2023), “Synthesis and in silico investigation of organoselenium-clubbed schiff bases as potential Mpro inhibitors for the SARS-CoV-2 replication”. The Life, 13(4), 912. https://doi.org/10.3390/life13040912
Shadid, M. Liu, Y., Veizaj, E., Huang, J., Johnson, J., and Kamel, A. (2018). Evaluation of rat brain subcellular fractions for the prediction of in vivo drug metabolism in brain. Drug Metabolism and Pharmacology, 33, S79. https://doi.org/10.1016/J.DMPK.2017.11.259
Szakács, G., Váradi, A., Ozvegy-Laczka, C., & Sarkadi, B. (2008). The role of ABC transporters in drug absorption, distribution, metabolism, excretion and toxicity (ADME-Tox). Drug Discovery Today, 13, 379–393.
Talevi, A. (2016). Computational approaches for innovative antiepileptic drug discovery. Expert Opinion in Drug Discovery, 11, 1001-1016
Xiong, G.L., Wu, Z. X., Yi, J. C., Fu, L., Yang, Z.J., Hsieh, C.H., et al. (2021). ADMETlab 2.0: an integrated online platform for accurate and comprehensive predictions of ADMET properties. Nucliec Acids Research, 49(W1), W5-W14. PMID:33893803.
Yadav, N., Ganie, S.A., Singh, B., Chhillar, A.K, & Yadav, S.S. (2019). Phytochemical constituents and ethnopharmacological properties of Ageratum conyzoides L. Phytotherapy Research, 1–16.
Yeon, S.S., Hye, S.B, Kazuo, O., & Kuk, H. (2005). Inhibitory effects of Isorhamnetin-3-O-b - D-glucoside from Salicornia herbacea on rat lens aldose reductase and sorbitol accumulation in streptozotocin-induced diabetic rat tissues. Biological and Pharmaceutical Bulletine, 28(5), 916-918. https://doi.org/10.1248/bpb.28.916.
Zhou, T., Cao, S., Su, P.C., Patel, R., Shah, D., Chokshi, H.B., et al. (2013). Hit identiļ¬cation and optimization in virtual screening: practical recommendations based upon a critical literature analysis. Journal of Medicinal Chemistry, 56, 6560-6572.