info@biomedres.us   +1 (502) 904-2126   One Westbrook Corporate Center, Suite 300, Westchester, IL 60154, USA   Site Map
ISSN: 2574 -1241

Impact Factor : 0.548

  Submit Manuscript

Research ArticleOpen Access

Synthesis and In Silico Analysis of Chalcone Derivatives as Potential Prostaglandin Synthetase Inhibitors Volume 55- Issue 2

Samuel J Bunu1,4*, Oyeintonbara Miediegha1, Ebisindor V Awala1, Deghinmotei Alfred-Ugbenbo2, Baba Haruna3 and Cyril O Usifoh4

  • 1Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmacy, Niger Delta University, Nigeria
  • 2Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Bayelsa Medical University, Nigeria
  • 3Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, University of Calabar, Nigeria
  • 4Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Benin, Nigeria

Received: February 14, 2024; Published: February 20, 2024

*Corresponding author: Samuel J Bunu, Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmacy, Niger Delta University, Wilberforce Island, Bayelsa State Nigeria

DOI: 10.26717/BJSTR.2024.55.008662

Abstract PDF

ABSTRACT

Nine derivatives of chalcones were successfully synthesized using the Claisen- Schmidt condensation reaction between different derivatives of benzaldehyde and acetophenone at low temperature in the presence of potassium hydroxide (KOH) and ethanol. The compounds were obtained in high yield. The percentage yield ranges from 90.38 – 27.68%, with sample B having the highest yield while sample I gave lowest yield. Also, the infra-red and nuclear magnetic resonance (FTIR and NMR) spectroscopic analysis shows distinct spectrum across all molecules, indicating the presence of unique functional groups and chemical environments. All the synthesized chalcones derivatives showed appreciable protein binding affinity against the COX-1 and COX-2 enzymes. The synthesized compounds had higher binding affinity against the COX-1 protein, compared to diclofenac and celecoxib that were used as standards. Sample A showed the highest affinity (-7.24 kcal/mol), while E showed the lowest affinity at -6.11 kcal/mol, higher than diclofenac (-5.46 kcal/mol) and comparable to celecoxib (-6.19 kcal/mol). For COX-2, Celecoxib (selective COX-2 blocker) showed highest binding affinity of -10.55 kcal/mol, while the test compounds B had -8.84 (highest among the test samples) and I (-8.69 kcal/mol (lowest affinity), with diclofenac having -8.49 kcal/mol respectively. Compounds E (4-methoxy- 4,6-diphenyl-2-thiopyrimidine) and B (para- chlorochalcone) from previous studies, displayed remarkable anti-inflammatory in an in-vivo animal model analysis. B showed the highest affinity against COX-2 and very high affinity towards COX-1 protein compared to the standard molecules. This shows that with adequate physiochemical and structural modifications, these compounds could serve as potential lead compounds in analgesic and anti-inflammatory pharmacology.

Keywords: Analgesic; Anti-Inflammatory; Chalcone Derivatives; Molecular Docking; Drug Design; In-Silico

Summary

Prostaglandins (PGs) are biochemical endogenous lipids with autacoid functions, synthesized in- vivo from arachidonic acid [1]. PGs and other similar physiologically active compounds are collectively known as metabolites of eicosanoids [2]. PGs have long been reported to have sustained homeostatic roles and facilitate many pathogenic mechanisms in inflammation, gastrointestinal tract [1], muscle contraction, blood clotting [3], ocular protection [4,5], and regulation of the circulatory system [6,7]. These lipids are produced via the action of the cyclo-oxygenase (COX-1 and COX-2) isoenzymes, and their biosynthesis is antagonized by the nonsteroidal anti-inflammatory drugs (NSAIDs) [1]. Some vitamins, including D3 (cholecalciferol), and K2 (menaquinone) are also known to inhibit the actions and biosynthesis of PGs [8-10]. PGs mainly take part in vasodilation, conception, luteolysis, menstruation, parturition, blood pressure reduction, control of sodium reabsorption by the kidney, etc [11]. Studies have shown that excessive concentrations of PGs induce diarrhea that accompanies medullary carcinoma of the thyroid or neural crest tumors and mediates several inflammatory responses [11], incoordinate hyperactivity of the uterine muscle leading to uterine ischemia, and menstrual cramps in women [12]. PG structural analogs like latanoprost, travoprost, and bimatoprost with antagonistic properties, are being used and well-tolerated for the reduction of intraocular pressure (IOP) in patients with primary open-angle glaucoma and ocular hypertension [7].

Previous reports have indicated that there are some correlations between high levels of PGs analog (PGFS) in tumors of the GI tract and the effectiveness of NSAIDs [13]. Thus, they can be used in the study, design, and discovery of antitumor agents. Cyclooxygenase 1 and 2 (COX-1 and COX-2) biologically transform arachidonic acid (AA) to prostaglandins H2 (PGH2), which is further biotransformed to various PGs, and other endogenous lipids like thromboxanes, leukotrienes, and hydroxyeicosateraenoic acids [14,15]. The names of these enzymes are derived from their catalytic cyclo-oxygenation that converts AA to prostaglandin G2 (PGG2), and peroxidation of PGG2 to PGH2, hence are also known as peroxidase enzymes as well [16]. The three COX isoforms, COX-1 COX-2, and COX-3, have been identified to share almost 60% amino acid sequence similarity but with much higher sequence homology in the catalytic sites (Figure 1) [17]. COX-1 is firmly expressed in many tissues, while COX-2 is strongly induced by various mitogens and plays imperative roles in many pathological conditions like inflammation [18,19]. There is also a COX-3 enzyme but reported to be not functional in humans. COX-3 isozyme is encoded by the same gene as COX-1, but COX-3 retains a particular nucleotide sequence (intron 94) that is not retained in COX-1 [20].

Figure 1

biomedres-openaccess-journal-bjstr

PGE2 increases gastric mucus secretion, uterus contraction (particularly during pregnancy), GI tract smooth muscle contraction, inhibition of lipolysis, autonomic neurotransmitters regulation, platelet response to agonists, and in-vivo atherothrombosis [21,22]. COX-1 enzyme regulates the baseline levels of PGs, while COX-2 synthesizes PGs via stimulation and significantly increases PGs levels during growth and inflammation, although both enzymes are located in the stomach, kidneys, and blood vessels [23]. Hence, inhibition of these agents is necessary for the optimal regulation of many biological functions, when in excessive amounts. NSAIDs inhibit the activities of COX-1 and COX-2 enzymes. There are non-selective (inhibits both COX-1 and COX-2), and COX-2 selective NSAIDs. These NSAIDs, while reducing inflammation caused by PGs, also inhibit platelet aggregation and increase the risk of GIT ulcers and intestinal bleeding [24]. COX-2 selective inhibitors promote thrombosis and increase the risk of heart attacks [25]. Due to these adverse reactions, coupled with the high etiology of vascular, and kidney disease complications, some COX-2 inhibitors are no longer in clinical use [26]. There are also reports that NSAIDs impair the production of erythropoietin, resulting in anaemia [27].

The long-term harmful effects of most NSAIDs outweigh the medical benefits. A study conducted a few years ago, observed a statistically significant increase in myocardial infarction incidence among patients on rofecoxib [28], and data from approved clinical trials, showed a significant relative risk of cardiovascular events that led to the global withdrawal of rofecoxib in 2004 [29]. Another study reported a significant increase in erectile dysfunction in men who frequently used NSAIDs [30]. NSAIDs are also associated with a doubled risk of heart failure in people who have not experienced cardiac disease in their lifetime [31]. Finally, the use of NSAIDs during late pregnancy can cause miscarriage [32], premature birth [33], constriction, and closure of fetal ductus arteriosus, leading to different blood-related congenital heart diseases in the fetus [34]. Acetaminophen, regarded as the safest, and well tolerated NSAID during pregnancy, was reported to cause male infertility in the fetus [35,36]. These advents call for the search for more effective, with minimal toxic molecules that can be used clinically to alleviate inflammatory conditions.

Chalcones chemically known as 1,3-diaryl-2-propen-1-ones, are flavonoids and isoflavonoids precursors, are chemical moieties present in many naturally compounds and are also prepared synthetically because of their convenient synthetic procedures [37]. Chalcone derivatives have been reported to possess antiproliferative [38], anti- inflammatory [39], antitumor [40], antimalarial [41], antibacterial [42], antiviral [43], antileishmanial [44], antifungal [45] properties, among others [46]. Molecular docking is a veritable tool used in the computational prediction of ligands and protein inhibitory affinity in the search for lead molecules [47], including characterized natural products [48]. Therefore, we conducted the synthesis and molecular docking of some chalones derivatives which biological properties were evaluated previously, that can serve as lead compounds in the design of anti- inflammatory and analgesic agents, especially as potential prostaglandin synthetase enzymes (COX-1 and COX-2) secretagogues inhibitors.

Method

Synthesis

Scheme 1: Synthesis of Methoxy, Halogenated and Aminated Chalcone Derivatives: All reagents used in the synthesis and other analysis were of analytical grades. The IR data was obtained from the FTIR-8400S instrument, Shimadzu global links, North America, while Nuclear Magnetic Resonance (NMR) experiment was performed on a 400 MHz instrument, obtained from Varian Inc. Palo Alto, California, USA. An equivalent weight of 10.6 g benzaldehyde and 12.0 g acetophenone were weighed, into a 100 ml flask having 25 mL EtOH, and stirred on ice (4-0℃). 20 ml of KOH (20%) was added with continuous stirring for 20 minutes and allowed to stand for 24 hours. Ice chips were added, and the mixture was titrated with 25 mL of 20% acetic acid (4-0℃). Precipitates were formed, filtered under suction and recrystallized with ethanol, dried, the percentage yield and melting point were determined. This procedure was repeated with different benzaldehyde derivatives, including para-methoxybenzaldehyde, para-chlorobenzaldehyde and para-dimethylaminobenzaldehyde, giving rise to various derivatives of chalone (Scheme 1).

Scheme 1

biomedres-openaccess-journal-bjstr

Scheme 2: Synthesis of 6-Diphenyl-2-Thiopyrimidine Chalcone Derivatives: From the initial 4-methoxy-chalone derivative obtained, an equivalent weight of 2.38 g 4- methoxy-chalone, 2.12 g sodium bicarbonate, and 1.52 g thiourea, were weighed into a flask having 30 mL DMSO. The mixture was refluxed under Nitrogen gas for about 2 hrs, using a Thin Layer Chromatographic plate to monitor the progress of the reaction. Water was added to the reaction medium at the end and was allowed to stand for 24 hours. Precipitates were formed, filtered under suction, the residues were recrystallized with diethyl-ether and petroleum spirit. The percentage yield and melting points of the crystals obtained were quantified, after drying (Scheme 2).

Scheme 2

biomedres-openaccess-journal-bjstr

Scheme 3: Synthesis of Epoxide Chalcone Derivatives: For the synthesis of epoxide derivatives, an equivalent weight of 5.16 g chalcone obtained previously was weighed into a beaker; 10 ml of 10% NaOH and 60 ml of MeOH were added respectively. The content of the beaker was dissolved with stirring via gentle heat, then 20 ml hydrogen peroxide H2O2) was added and stirred for 30 minutes. 5 ml of 10% acetic acid was used to acidify the medium. The resultant product was collected, and recrystallized with MeOH, filtered and dried. The percentage yield and melting points were respectively determined. This procedure was repeated with para-methoxychalcone, para-chlorochalcone, para- dimethylaminochalone, respectively, leading to the production of chalcone epoxide derivatives (Scheme 3).

Scheme 3

biomedres-openaccess-journal-bjstr

Molecular Docking

Molecular modeling and docking simulations of the binding protein and synthesized ligands were done using the Maestro software of OPLS3, 2018 Force field [49], and Pymol software [50]. The docking parameters and affinity were compared with the previously reported pharmacological profile of the chalcone derivatives. The human COX-1 crystal structure protein (6Y3C) [51], and 1PXX (COX- 2 crystal structure with diclofenac bound to the cyclooxygenase active site) [52], were obtained from the PDB website, and modeled with the Pymol and D3Pocket webserver [53,54], to obtain all possible binding pockets and utilize one(s) with the highest affinity using diclofenac and Celecoxib (a selective COX-2 inhibitor) as the standard molecules.

A. (E)-Chalcone.
B. Para-chlorochalcone [(E)-3-(4-chlorophenyl)-1-phenylprop- 2-en-1-one]
C. Para-methoxychalcone [(E)-3-(4-methoxyphenyl)-1-phenylprop- 2-en-1-one]
D. Para-dimethylaminochalcone [(E)-3-(4-(dimethylamino) phenyl)-1-phenylprop-2-en-1-one]
E. Para-methoxy-4,6-diphenyl-2-thiopyrimidine[4-(4-methoxyphenyl)- 6-phenyl-5,6 dihydropyrimidine 2(1H)-thione]
F. Chalcone-epoxide [phenyl(3-phenyloxiran-2-yl) methadone]
G. Para-chlorochalcone-epoxide [(3-(4-chlorophenyl) oxiran- 2-yl) (phenyl)methanone]
H. Para-methoxychalcone-epoxide [(3-(4-methoxyphenyl) oxiran- 2-yl) (phenyl)methanone]
I. Para-dimethylaminochalcone-epoxide[(3-(4-(dimethylamino) phenyl)oxiran-2-yl)(phenyl)methanone]

Discussion

The compounds were obtained in high yield after the synthetic processes (Schemes 1 & 2). The percentage yields of the compound ranged from 27.68 – 90.38%, with sample B having the highest yield while sample I gave lowest synthetic yield. Also, the spectroscopic (FTIR and NMR) analysis shows distinct spectrum across all molecules, indicating the presence of unique functional groups and chemical environments (Table 1). All the synthesized chalcones derivatives showed appreciable protein binding affinity against the COX-1 and COX-2 enzymes. Compared to the standard drugs, better protein-ligand affinity was observed with COX-2 enzyme. Diclofenac is known to inhibit both COX-1 and COX-2 enzymes [55]. The computational experiment showed that, some of the synthesized compounds had higher binding affinity against the COX-1 protein than both diclofenac and celecoxib. Compound A showed the highest affinity (-7.24 kcal/mol), while other compounds had affinity level of -7.21(C), -7.16 (B), -7.14 (I), -7.10 (D), -7.00 (H), -6.92 (G), -6.88 (F), and -6.11 kcal/mol(E), respectively. Whereas, the standard compounds (diclofenac and celecoxib) had -5.46 and -6.19 kcal/mol, respectively.

Table 1: Physiochemical and elemental analysis of Chalcone derivatives.

biomedres-openaccess-journal-bjstr

The protein-ligand interactions showed the actual protein residues in the COX-1 protein that compounds bound with (Figure 2). Also, the binding pocket and pose of the compound with highest affinity showed how it is well fitted into the protein pocket (Figure 3). Celecoxib, a selective COX-2 inhibitor showed highest binding affinity of -10.55 kcal/mol, while the test compounds had -8.55 (A), -8.84 (B) -8.50 (C), -8.79 (D), -8.24 (E) -8.13 (F), -8.50 (G), -8.22 (H), -8.69 (I), and diclofenac had -8.49 kcal/mol respectively (Table 2). The binding interactions of all molecules are shown in Figure 4, while binding poses of the molecules with the highest affinity is illustrated in Figure 5. Compounds E (4-methoxy-4,6-diphenyl-2-thiopyrimidine) and B (para-chlorochalcone) from previous studies, displayed remarkable anti-inflammatory in an in-vivo analysis using animal model [39]. E also showed the appreciable affinity against COX-1 protein more than the standard compounds, while lower affinity was observed against COX-2 protein. For compound B, it showed the highest affinity against COX-2 and very high affinity towards COX-1 protein compared to the standard molecules used in the analysis. This shows that with adequate physiochemical and structural modifications, these compounds could serve as potential lead compounds in analgesic and anti-inflammatory pharmacology, as pain and inflammation are associated with these enzymes in the biological system [16].

Figure 2

biomedres-openaccess-journal-bjstr

Figure 3

biomedres-openaccess-journal-bjstr

Figure 4

biomedres-openaccess-journal-bjstr

Figure 5

biomedres-openaccess-journal-bjstr

Table 2: Structural features and Molecular docking of the synthesized compounds.

biomedres-openaccess-journal-bjstr

Conclusion

Analgesic and anti-inflammatory agents are used in the prevention of all kinds of pains, ranging from minor headaches to severe post-operative pains. The search for newer agents due to poor tolerability, adverse reactions and affordability of the existing ones is the focus of contemporary drug design and development. The compounds showed highly promising results in both in-vivo and computational molecular docking studies. Hence, the compounds reported in this study could be utilized and further modified structurally and physiochemically to achieve better analgesic and anti-inflammatory properties.

Funding

This work was partially funded by the Tertiary Education Trust Fund (TETFund) Nigeria.

Acknowledgment

The authors sincerely acknowledge the contribution of Prof. James O. Oluwdiya, Prof. Benjamin U. Ebeshi, all staff of the Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmacy, Niger Delta University, Wilberforce Island, Bayelsa State, and Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Benin, Nigeria.

Authors Contribution

SBJ: in silico studies, data analysis, manuscript original draft. OM: data curation, manuscript review. EVA: synthetic studies, preliminary in-vivo studies. DA: manuscript critical review. BA: supervision, conception, validation. COU: supervision, spectroscopic analysis, data interpretation, manuscript critical review and approval.

Conflict of Interests

The authors declare no conflict of interest.

References

  1. Ricciotti E, FitzGerald GA (2011) Prostaglandins and inflammation. Arterioscler Thromb Vasc Biol 31(5): 986-1000.
  2. Imig J D (2020) Eicosanoid blood vessel regulation in physiological and pathological states. Clin Sci (Lond) 134(20): 2707-2727.
  3. Komoto J, Yamada T, Watanabe K, Takusagawa F (2004) Crystal structure of human prostaglandin F synthase (AKR1C3). Biochemistry 43(8): 2188-2198.
  4. Kass MA, Podos SM, Moses RA, Becker B (1972) Prostaglandin E 1 and aqueous humor dynamics. Invest Ophthalmol 11(12): 1022-1027.
  5. Nkechi J Onyeukwu, SJB, Tologbonse A Adedoyinv, Ngozi A Onwuka (2023) Comparative Analysis of Petrochemicals Effects on Ocular Cavity: Prevalence and Pharmaceutical Interventions in the Niger Delta, Nigeria. Mathews Journal of Pharmaceutical Science 7(2): 1-16.
  6. Eakins KE (1977) Prostaglandin and non-prostaglandin mediated breeakdown of the blood-aqueous barrier. Exp Eye Res 25: 483-498.
  7. Alexander CL, Miller SJ, Abel SR (2002) Prostaglandin analog treatment of glaucoma and ocular hypertension. Ann Pharmacother 36(3): 504-511.
  8. Kieronska Rudek A, Kij A, Kaczara P, Tworzydlo A, Napiorkowski M, et al. (2021) Exogenous Vitamins K Exert Anti-Inflammatory Effects Dissociated from Their Role as Substrates for Synthesis of Endogenous MK-4 in Murine Macrophages Cell Line. Cells 10(7).
  9. Krishnan A V, Srinivas S, Feldman D (2009) Inhibition of prostaglandin synthesis and actions contributes to the beneficial effects of calcitriol in prostate cancer. Dermatoendocrinol 1(1): 7-11.
  10. Koshihara Y, Hoshi K, Shiraki M (1993) Vitamin K2 (menatetrenone) inhibits prostaglandin synthesis in cultured human osteoblast-like periosteal cells by inhibiting prostaglandin H synthase activity. Biochem Pharmacol 46(8): 1355-1362.
  11. Jones R L (1972) Functions of prostaglandins. Pathobiol Annu 2: 359-380.
  12. Dawood MY (1981) Dysmenorrhoea and prostaglandins: pharmacological and therapeutic considerations. Drugs 22(1): 42-56.
  13. Komoto J, Yamada T, Watanabe K, Woodward D F, Takusagawa F (2006) Prostaglandin F2alpha formation from prostaglandin H2 by prostaglandin F synthase (PGFS): crystal structure of PGFS containing bimatoprost. Biochemistry 45(7): 1987-1996.
  14. Narumiya S, Sugimoto Y, Ushikubi F (1999) Prostanoid receptors: structures, properties, and functions. Physiol Rev 79(4): 1193-1226.
  15. Regan J W (2003) EP2 and EP4 prostanoid receptor signaling. Life Sci 74(2-3): 143-153.
  16. Marnett LJ (2000) Cyclooxygenase mechanisms. Curr Opin Chem Biol 4(5): 545-552.
  17. Smith WL, DeWitt DL, Garavito RM (2000) Cyclooxygenases: structural, cellular, and molecular biology. Annu Rev Biochem 69: 145-182.
  18. Turini ME, DuBois RN (2002) Cyclooxygenase-2: a therapeutic target. Annu Rev Med 53: 35-57.
  19. Vane JR, Bakhle Y S, Botting R M (1998) Cyclooxygenases 1 and 2. Annu Rev Pharmacol Toxicol 38: 97-120.
  20. Chandrasekharan N V, Dai H, Roos KL, Evanson N K, Tomsik J, et al. (2002) COX-3, a cyclooxygenase-1 variant inhibited by acetaminophen and other analgesic/antipyretic drugs: cloning, structure, and expression. Proc Natl Acad Sci U S A 99(21): 13926-13931.
  21. Fabre JE, Nguyen M, Athirakul K, Coggins K, McNeish J D, et al. (2001) Activation of the murine EP3 receptor for PGE2 inhibits cAMP production and promotes platelet aggregation. J Clin Invest 107(5): 603-610.
  22. Gross S, Tilly P, Hentsch D, Vonesch JL, Fabre JE (2007) Vascular wall-produced prostaglandin E2 exacerbates arterial thrombosis and atherothrombosis through platelet EP3 receptors. J Exp Med 204(2): 311-320.
  23. Ke J, Yang Y, Che Q, Jiang F, Wang H, et al. (2016) Prostaglandin E2 (PGE2) promotes proliferation and invasion by enhancing SUMO- 1 activity via EP4 receptor in endometrial cancer. Tumour Biol 37(9): 12203-12211.
  24. Rostom A, Dube C, Wells G, Tugwell P, Welch V, et al. (2002) Prevention of NSAID-induced gastroduodenal ulcers. Cochrane Database Syst Rev (4): CD002296.
  25. Day RO, Graham GG (2004) The vascular effects of COX-2 selective inhibitors. Australian Prescriber 27(6): 142-145.
  26. Brater DC, Harris C, Redfern JS, Gertz BJ (2001) Renal effects of COX-2-selective inhibitors. Am J Nephrol 21(1): 1-15.
  27. Bleumink GS, Feenstra J, Sturkenboom MC, Stricker BH (2003) Nonsteroidal anti- inflammatory drugs and heart failure. Drugs 63(6): 525-534.
  28. Baron J A, Sandler RS, Bresalier RS, Lanas A, Morton DG, et al. (2008) Cardiovascular events associated with rofecoxib: final analysis of the APPROVe trial. Lancet 372(9651): 1756-1764.
  29. Sibbald B (2004) Rofecoxib (Vioxx) voluntarily withdrawn from market. Canadian Medical Association Journal 171(9): 1027-1028.
  30. Shiri R, Koskimaki J, Hakkinen J, Tammela T L, Auvinen A, et al. (2006) Effect of nonsteroidal anti-inflammatory drug use on the incidence of erectile dysfunction. J Urol 175(5): 1812-1815.
  31. Coxib traditional NTC, Bhala N, Emberson J, Merhi A, Abramson S, et al. (2013) Vascular and upper gastrointestinal effects of non-steroidal anti-inflammatory drugs: meta-analyses of individual participant data from randomised trials. Lancet 382(9894): 769-779.
  32. Nakhai Pour HR, Broy P, Sheehy O, Berard A (2011) Use of nonaspirin nonsteroidal anti- inflammatory drugs during pregnancy and the risk of spontaneous abortion. CMAJ 183(15): 1713-1720.
  33. Ostensen ME, Skomsvoll JF (2004) Anti-inflammatory pharmacotherapy during pregnancy. Expert Opin Pharmacother 5(3): 571-580.
  34. Koren G, Florescu A, Costei AM, Boskovic R, Moretti ME (2006) Nonsteroidal antiinflammatory drugs during third trimester and the risk of premature closure of the ductus arteriosus: a meta-analysis. Ann Pharmacother 40(5): 824-829.
  35. Graham GG, Scott KF, Day RO (2005) Tolerability of paracetamol. Drug Saf 28(3): 227-240.
  36. Kristensen DM, Hass U, Lesne L, Lottrup G, Jacobsen P R, et al. (2011) Intrauterine exposure to mild analgesics is a risk factor for development of male reproductive disorders in human and rat. Hum Reprod 26(1): 235-244.
  37. Ouyang Y, Li J, Chen X, Fu X, Sun S, et al. (2021) Chalcone Derivatives: Role in Anticancer Therapy. Biomolecules 11(6): 894.
  38. Burmaoglu S, Aktas Anil D, Gobek A, Kilic D, Yetkin D, et al. (2022) Design, synthesis and antiproliferative activity evaluation of fluorine-containing chalcone derivatives. J Biomol Struct Dyn 40(8): 3525-3550.
  39. Awala E Victoria, BSJ, Baba Haruna, Oluwadiya J Oluwadiya (2019) Synthesis, Characterization, Antimicrobial and Anti-Inflammatory Properties of 4-Methoxy, 4, 6- Dipheny L-2-Thiopyrimidine and Epoxide Derivatives of Chalcones. Scholars Academic Journal of Pharmacy 8(8): 436-442.
  40. Gao F, Huang G, Xiao J (2020) Chalcone hybrids as potential anticancer agents: Current development, mechanism of action, and structure-activity relationship. Med Res Rev 40(5): 2049-2084.
  41. Cheng P, Yang L, Huang X, Wang X, Gong M (2020) Chalcone hybrids and their antimalarial activity. Arch Pharm (Weinheim) 353(4): e1900350.
  42. Pereira D, Duraes F, Szemeredi N, Freitas da Silva J, Pinto E, et al. (2022) New Chalcone-Triazole Hybrids with Promising Antimicrobial Activity in Multidrug Resistance Strains. Int J Mol Sci 23(22): 14291.
  43. Wang L, Yang R, Yuan B, Liu Y, Liu C (2015) The antiviral and antimicrobial activities of licorice, a widely used Chinese herb. Acta Pharm Sin B 5(4): 310-315.
  44. De Mello MVP, Abrahim Vieira BA, Domingos TFS, De Jesus JB, de Sousa ACC et al. (2018) A comprehensive review of chalcone derivatives as antileishmanial agents. Eur J Med Chem 150: 920-929.
  45. Bunu Samuel Jacob, AEV, Eboh Darlington Deboh (2020) Preparation and Antifungal Properties of Chalcone and Halogenated Derivatives Saudi Journal of Medical and Pharmaceutical Sciences 6(4): 379-389.
  46. Elkanzi NAA, Hrichi H, Alolayan RA, Derafa W, Zahou FM, et al. (2022) Synthesis of Chalcones Derivatives and Their Biological Activities: A Review. ACS Omega 7(32): 27769-27786.
  47. Samuel JBD Alfred Ugbenbo, Miediegha Oyeintonbara, Haruna Baba (2023) Characterization and Molecular Docking of Cinnamic acid derivatives: potential inhibitors of cyclo-oxygenase enzymes. Innovare Journal of Life Sciences 11(2023): 41-46.
  48. Azibanasamesa DC OF Arueniobebh, Samuel J Bunu, Raji O Rafiu, Ekarika C Johnson, Emmanuel I Etim (2024) Spectroscopic Analysis, Aphrodisiac Potential of Carapa Procera Stem Bark Extract in Male Wistar Rats and In Silico Studies of Hexadecanoic and Oleic Acids on Phosphodiesterase-5 and Adenylcyclase Enzymes. Biomed J Sci & Tech Res 54(5): 46343-46356.
  49. Salam NK, Adzhigirey M, Sherman W, Pearlman DA (2014) Structure-based approach to the prediction of disulfide bonds in proteins. Protein Eng Des Sel 27(10): 365-374.
  50. Seeliger D, de Groot BL (2010) Ligand docking and binding site analysis with PyMOL and Autodock/Vina. J Comput Aided Mol Des 24(5): 417-422.
  51. Miciaccia M, Belviso BD, Iaselli M, Cingolani G, Ferorelli S, et al. (2021) Three-dimensional structure of human cyclooxygenase (hCOX)-1. Sci Rep 11(1): 4312.
  52. Rowlinson SW, Kiefer JR, Prusakiewicz JJ, Pawlitz JL, Kozak KR, et al. (2003) A novel mechanism of cyclooxygenase-2 inhibition involving interactions with Ser-530 and Tyr-385. J Biol Chem 278(46): 45763-45769.
  53. Chen Z, Zhang X, Peng C, Wang J, Xu Z, et al. (2019) D3Pockets: A Method and Web Server for Systematic Analysis of Protein Pocket Dynamics. J Chem Inf Model 59(8): 3353-3358.
  54. Shi Y, Zhang X, Mu K, Peng C, Zhu Z, et al. (2020) D3Targets-2019-nCoV: a webserver for predicting drug targets and for multi-target and multi- site based virtual screening against COVID-19. Acta Pharm Sin B 10(7): 1239-1248.
  55. Stiller CO, Hjemdahl P (2022) Lessons from 20 years with COX-2 inhibitors: Importance of dose-response considerations and fair play in comparative trials. J Intern Med 292(4): 557-574.