CC BY 4.0 · Pharmaceutical Fronts 2022; 04(04): e250-e266
DOI: 10.1055/s-0042-1759688
Original Article

Molecular Docking, Drug-Likeness Analysis, In Silico Pharmacokinetics, and Toxicity Studies of p-Nitrophenyl Hydrazones as Anti-inflammatory Compounds against COX-2, 5-LOX, and H+/K+ ATPase

Sodeeq Babalola
1   Department of Pharmaceutical and Medicinal Chemistry, Ahmadu Bello University, Zaria, Nigeria
,
Nosakhare Igie
2   Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, Texas, United States
,
Isaiah Odeyemi
2   Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, Texas, United States
› Author Affiliations

Abstract

Nonsteroidal anti-inflammatory drugs (NSAIDs) and coxibs are traditional medicines for the treatment of inflammation, yet associated with serious side effects. Hence, the need for discovering novel compounds with valuable clinical benefits is of great importance. In this study, 18 derivatives of p-nitrophenyl hydrazones were docked against COX-2, 5-LOX, and H+/K+ ATPase, followed by predicting their drug-likeness and absorption, distribution, metabolism, and excretion (ADME) properties. From the docking analysis, 1-(4-nitrophenyl)-2-[(3,4,5-trimethoxyphenyl)methylidene]hydrazine (3), 4-hydroxy-2-methyl-6-[(2-(4-nitrophenyl)hydraz-1-ylidene)methyl]thiochroman-1,1-dioxide (6), 4-methoxy-2-methyl-6-[(2-(4-nitrophenyl)hydraz-1-ylidene)methyl]thiochroman-1,1-dioxide (8), 2-methyl-6-[(2-(4-nitrophenyl)hydraz-1-ylidene)methyl]-4-(trifluoromethyl)thiochroman-1,1-dioxide (11), 4-[(2-(4-nitrophenyl)hydraz-1-ylidene)methyl]benzenesulfonamide (13), 4-[(2-(4-nitrophenyl)hydraz-1-ylidene)methyl]-3-(trifluoromethyl)benzenesulfonamide (14), 5-methyl-6-{4-[(2-(4-nitrophenyl)hydraz-1-ylidene)methyl]phenyl}-2,3,4,5-tetrahydropyridazin-3-ol (16), and 5-methyl-6-{4-[(2-(4-nitrophenyl)hydraz-1-ylidene)methyl]phenyl}-4,5-dihydropyridazin-3(2H)-one (17) showed promise as potent multi-target inhibitors of COX-2, 5-LOX, and H+/K+ ATPase. These compounds are less COX-2 selective than the control (celecoxib). “Drug-likeness” analysis passed Lipinski's, Egan's, Veber's, Muegge's, and Ghose's rules. The compounds also passed Pfizer and GSK rules, as well as golden triangle's rule for identification of potent and metabolically stable drugs. The pharmacokinetic profiles of the compounds were excellent, safe, and compliant with their potential anti-inflammatory activity. The results of the study can be used for future optimization of those derivatives for better molecular interactions against COX-2, 5-LOX, and H+/K+ ATPase, and inflammation-effective inhibition.

Supplementary Material



Publication History

Received: 09 June 2022

Accepted: 03 November 2022

Article published online:
27 December 2022

© 2022. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Abdelgawad MA, Labib MB, Abdel-Latif M. Pyrazole-hydrazone derivatives as anti-inflammatory agents: design, synthesis, biological evaluation, COX-1,2/5-LOX inhibition and docking study. Bioorg Chem 2017; 74: 212-220
  • 2 Ashley NT, Weil ZM, Nelson RJ. Inflammation: mechanisms, costs, and natural variation. Annu Rev Ecol Evol Syst 2012; 43: 385-406
  • 3 Gökçe M, Utku S, Küpeli E. Synthesis and analgesic and anti-inflammatory activities 6-substituted-3(2H)-pyridazinone-2-acetyl-2-(p-substituted/nonsubstituted benzal)hydrazone derivatives. Eur J Med Chem 2009; 44 (09) 3760-3764
  • 4 Wang SM, Zha GF, Rakesh KP. et al. Synthesis of benzo[d]thiazole-hydrazone analogues: molecular docking and SAR studies of potential H+/K+ ATPase inhibitors and anti-inflammatory agents. MedChemComm 2017; 8 (06) 1173-1189
  • 5 Nguyen HT, Vu TY, Chandi V, Polimati H, Tatipamula VB. Dual COX and 5-LOX inhibition by clerodane diterpenes from seeds of Polyalthia longifolia (Sonn.) Thwaites. Sci Rep 2020; 10 (01) 15965
  • 6 Kumar N, Chauhan L. Analgesic and anti-inflammatory potential of hydrazones. J Chem Pharm Res 2014; 6: 916-934
  • 7 Asif M, Husain A. Analgesic, anti-inflammatory, and antiplatelet profile of hydrazones containing synthetic molecules. J Appl Chem 2013; 2013: 247203
  • 8 Hiroyuki Y, Yuichiro S, Masato F. Chronic inflammatory disease therapeutic agent. JP Patent 2012046453A. March, 2012
  • 9 Kulkarni RG, Achaiah G, Sastry GN. Novel targets for antiinflammatory and antiarthritic agents. Curr Pharm Des 2006; 12 (19) 2437-2454
  • 10 Ivanenkov YA, Zagribelnyy BA, Aladinskiy VA. Are we opening the door to a new era of medicinal chemistry or being collapsed to a chemical singularity?. J Med Chem 2019; 62 (22) 10026-10043
  • 11 Xiong G, Wu Z, Yi J. et al. ADMETlab 2.0: an integrated online platform for accurate and comprehensive predictions of ADMET properties. Nucleic Acids Res 2021; 49 (W1): W5-W14
  • 12 Liu X, Wright M, Hop CE. Rational use of plasma protein and tissue binding data in drug design. J Med Chem 2014; 57 (20) 8238-8248
  • 13 Cecchelli R, Berezowski V, Lundquist S. et al. Modelling of the blood-brain barrier in drug discovery and development. Nat Rev Drug Discov 2007; 6 (08) 650-661
  • 14 Stanley LA. Part VI: Basic animal anatomy and physiology. In: Badal Mccreath S, Delgoda R. eds. Pharmacognosy. 1st ed.. Amsterdam: Elsevier; 2017: 528-545
  • 15 Brambilla G, Martelli A. Genotoxicity and carcinogenicity studies of analgesics, anti-inflammatory drugs and antipyretics. Pharmacol Res 2009; 60 (01) 1-17
  • 16 Yoshioka S, Ohno N, Miura T, Adachi Y, Yadomae T. Immunotoxicity of soluble beta-glucans induced by indomethacin treatment. FEMS Immunol Med Microbiol 1998; 21 (03) 171-179
  • 17 Takahashi H, Ohno N, Adachi Y, Yadomae T. Association of immunological disorders in lethal side effect of NSAIDs on beta-glucan-administered mice. FEMS Immunol Med Microbiol 2001; 31 (01) 1-14
  • 18 Kuboyama N, Fujii A. Mutagenicity of analgesics, their derivatives, and anti-inflammatory drugs with S-9 mix of several animal species. J Nihon Univ Sch Dent 1992; 34 (03) 183-195
  • 19 Tomisato W, Tsutsumi S, Rokutan K, Tsuchiya T, Mizushima T. NSAIDs induce both necrosis and apoptosis in guinea pig gastric mucosal cells in primary culture. Am J Physiol Gastrointest Liver Physiol 2001; 281 (04) G1092-G1100
  • 20 Reuter H, Tarr Gs. Review of the safety of nonsteroidal anti-inflammatory drugs and selective cyclo-oxygenase-2 inhibitors. S Afr Fam Pract 2019; 57: 18-22
  • 21 Cryer B, Dubois A. The advent of highly selective inhibitors of cyclooxygenase–a review. Prostaglandins Other Lipid Mediat 1998; 56 (5–6): 341-361
  • 22 Stevenson DD, Sanchez-Borges M, Szczeklik A. Classification of allergic and pseudoallergic reactions to drugs that inhibit cyclooxygenase enzymes. Ann Allergy Asthma Immunol 2001; 87 (03) 177-180
  • 23 Namazy JA, Simon RA. Sensitivity to nonsteroidal anti-inflammatory drugs. Ann Allergy Asthma Immunol 2002; 89 (06) 542-550 , quiz 550, 605
  • 24 Tian Y, Ouyang N, Thomas PE. Regulation of xenobiotic sensor PXR and AhR by NF-B and its roles in xenobiotic detoxification and inflammation-associated carcinogenesis. In: McQueen CA. ed. Comprehensive Toxicology. 2nd ed.. Amsterdam: Elsevier Science; 2010: 579-585
  • 25 Patergnani S, Vitto VAM, Pinton P, Rimessi A. Mitochondrial stress responses and “mito-inflammation” in cystic fibrosis. Front Pharmacol 2020; 11: 581114
  • 26 Environmental Protection Agency. Persistent bioaccumulative toxic (PBT) chemicals; lowering of reporting thresholds for certain PBT chemicals; addition of certain PBT chemicals; community right-to-know toxic chemical reporting. Fed Regist 1999; 64 (209) 58666-58753
  • 27 Babalola S, Igie N, Odeyemi I. Structure-based discovery of multitarget directed anti-inflammatory p-nitrophenyl hydrazones; molecular docking, drug-likeness, in-silico pharmacokinetics, and toxicity studies. ChemRxiv. Cambridge: Cambridge Open Engage; 2022
  • 28 Babalola S, Igie N, Odeyemi I, Idris AY, Sanni YM, Hamza AN. In-vivo anti-inflammatory activity studies of some p-nitrophenyl hydrazones. Discovery (Read) 2022; 58 (319) 698-706
  • 29 Shin JM, Munson K, Vagin O, Sachs G. The gastric HK-ATPase: structure, function, and inhibition. Pflugers Arch 2009; 457 (03) 609-622
  • 30 Jin X, Luong TL, Reese N. et al. Comparison of MDCK-MDR1 and Caco-2 cell based permeability assays for anti-malarial drug screening and drug investigations. J Pharmacol Toxicol Methods 2014; 70 (02) 188-194
  • 31 Smith DA, Di L, Kerns EH. The effect of plasma protein binding on in vivo efficacy: misconceptions in drug discovery. Nat Rev Drug Discov 2010; 9 (12) 929-939