Skip to content
BY 4.0 license Open Access Published by De Gruyter March 23, 2022

Cyclization of N-acetyl derivative: Novel synthesis – azoles and azines, antimicrobial activities, and computational studies

  • Eman O. Hamed , Mohamed G. Assy , Nabil H. Ouf , Doaa A. Elsayed and Magda H. Abdellattif EMAIL logo

Abstract

2-Pyridone is considered as one of the most famous efficient pharmaceutical compounds. Many approaches were discovered to synthesize 2-pyridone. In this present research, chloroacetylation of benzylamine at simple conditions, EtONa/EtCOONa produced N-benzyl-2-chloroacetamide 2. Compound 2 was allowed to react with different reagents. These reagents are acetylacetone, ethyl cyanoacetate, ethyl acetoacetate, and diethyl malonate, creating 2-pyridone derivatives with a good yield. The structures of the prepared compounds were elucidated by spectral data (IR, 1HNMR, and 13CNMR). The synthesized compound was tested for its antimicrobial activity against the Gram-positive (Staphylococcus aureus) and the Gram-negative (Escherichia coli) bacteria. In addition, the antifungal activities of the compounds were tested against two fungi (Candida albicans and Aspergillus flavus). Molecular docking studies were applied using the Autodock vina method. Theoretical methods prove all the experimental results by using molecular docking using Autodock vina and by ADEMT studies. The docking results represent that compound 20 had the best docking free energy, and it is the effective compound toward the selected bacterial and fungal proteins. ADME studies showed that the only compound 18 could cross the blood–brain barrier, and compound 15 was predicted to be soluble.

1 Introduction

Cyclization of α-halogenacetamides is a building block for synthesizing several organic compounds, which attracted the attention of many researchers due to their importance in various fields [1,2,3]. While some derivatives occur naturally, 2-pyridone does not [4,5]. 2-Pyridone has two forms of tautomerism (keto and enol forms), expressed as lactam–lactim tautomerism. In this type of tautomerism, the H+ on the nitrogen atom can be attached to the oxygen atom to obtain the second tautomer. In the solid state, 2-pyridone exists in the keto form, where the IR analysis indicated the presence of the ketone group. However, in the solution state, 2-pyridone tautomerization depends on the polarity of the solvent, wherein polar solvents exist in ketone form, while nonpolar solvents present as the hydroxyl form [6]. The dimerization of 2-pyridone and 2-hydroxy pyridine underwent through hydrogen bond [7].

α-Pyridone derivatives have wide and important applications in many fields like industry, biology, and medicine. They are used as anticancer [8,9], anti-inflammatory [10,11], antimicrobial, antifungal [12,13,14,15], pharmaceutical [16], antiviral [17,18], anti-oxidant [19], dyes [20,21,22,23,24], fluorescents [25], and natural products [26,27]. In addition, 2-pyridone has catalytic activity, and hence, it can be used as a catalyst in proton-dependent reactions like aminolysis of esters [28,29,30] and its uses as a ligand in coordination chemistry [31,32]. Therefore, these animated researchers prepare 2-pyridone and its derivatives through cyclic and condensation of an acyclic system [33,34,35,36,37,38,39,40]. Thus, in this work, the preparation and properties of some 2-pyridone derivatives are presented. Computational studies involving molecular docking by auto dock vina were used to predict and prove the biological studies. ADMET studies indicated the prediction and validation of the activity of the synthesized compounds. The use of computational studies is one of the modern techniques that help describe the pharmacological properties of the synthesized compounds. The consequence of the present research strengthens the applicability of these compounds as encouraging anticancer and antibacterial drugs that could help the medicinal chemists and pharmaceuticals further design and synthesize more effective drug candidates [41,42,43,44].

2 Results and discussion

2.1 Chemistry

Chloroacetylation of benzylamine furnished multifunction product 2 (Scheme 1). The stretching frequency of 2 was observed at 3,271, 1,641, and 1,546 cm−1 for N–H, C═O, and C═C, respectively. The exchangeable downfield signal of N–H was shown at 8.70 ppm. The synthetic strategy of producing a new pyridone system depends on the reaction of active methylene with 1 followed by intramolecular cyclization involving the condensation of the ester carbonyl with nucleophilic benzylic carbon in a basic medium. Thus, diethyl malonate underwent alkylation, cyclization followed by hydrolysis, subsequent CO2 evolution, and enolization to furnish 6 (Scheme 1). IR spectrum led to peaks at 3,274, 1,644, and 1,549 for O–H, C═N, and C═C, respectively. O–H appeared at 13.2 and 15.14 ppm.

Scheme 1 
                  Alkylation followed by cyclization of 2 with active methylene.
Scheme 1

Alkylation followed by cyclization of 2 with active methylene.

Acetyl acetone underwent alkylation by using 2 in a basic medium followed by cyclodehydration producing pyridone derivative 8. N–H, C═O, and C═C stretching frequency were observed at 3,275, 1,645, and 1,549 cm−1, respectively. The N–H signal was detected at 11.09 ppm, and the carbonyl carbon signal was detected at 139.29 and 166.43.

Ethyl cyanoacetate underwent SN reaction with 2, cyclization, iminohydrolysis, dehydrogenation followed by ester hydrolysis, and subsequent decarboxylation leading to 12 (Scheme 2). IR spectrum led to C═O and C═N, while there is no N–H signal.

Scheme 2 
                  Azole and azine generation from acylated product 2.
Scheme 2

Azole and azine generation from acylated product 2.

As depicted in Scheme 2, cyclo-condensation of 2 and ethyl acetoacetate furnished pyridone derivative 15. IR peaks presented at 3,275, 1,644, and 1,548 cm−1 due to O–H, C═N, and C═C, respectively. The O–H downfield signal was observed at 12.2 ppm. The imidazole cyclization 16 was observed by using thiourea as a cyclizing agent. IR spectrum showed peaks at 3,270, 1,641, and 1,311 for N–H, CN, and C═S, respectively. Intermolecular cyclization of 2 and benzaldehyde led to benzylic condensation followed by Michael’s addition. IR spectrum led to N–H, C═O, and C═C function, the N–H signal was observed at 8.62 ppm, and carbonyl carbon was located at 134.01.

Subjecting 2 to added to heteroallene electrophilic carbon proved piperazine cyclization led to 20 (Scheme 3). N–H, CO, and C═S functions were observed at 3,272, 1,638, and 1,226 cm−1, respectively. N–H signal was observed at 8.79 ppm.

Scheme 3 
                  Cyclization of compound 2 with benzoyl isothiocyanate.
Scheme 3

Cyclization of compound 2 with benzoyl isothiocyanate.

2.2 Biology

The biological activity of the produced compounds was studied for their antimicrobial effects against the Gram-positive (Staphylococcus aureus) and the Gram-negative (Escherichia coli) bacteria. In addition, antifungal actions of the compounds were tested against two fungi (Candida albicans and Aspergillus flavus).

Table 1 shows that compounds 15, 18, and 20 have inhibitory effects on S. aureus, E. coli, A. flavus, C. albicans strains [41,42]. The antimicrobial activity of the tested compounds was determined using a modified Kirby-Bauer disc diffusion method [42].

Table 1

In vitro antibacterial and antifungal screening of the newly synthesized compounds

Sample Inhibition zone diameter (mm/mg sample)
Bacterial species Fungal species
G+ G
Staphylococcus aureus Escherichia coli Aspergillus flavus Candida albicans
Control: DMSO 0 0 0 0
6 0.0 0.0 0.0 0.0
8 9 0 0 0
15 10 12 0 0
18 10 10 0 0
20 11 15 10 26
Ampicillin Antibacterial agent 21 25
Amphotericin B 17 21
Antifungal agent

Briefly, 100 µL of the test bacteria/fungi were grown in 10 mL of fresh media until they reached a count of approximately 108 cells/mL for bacteria 105 cells/mL for fungi [43].

2.3 Computational studies

2.3.1 Analyses of the docked complexes and ADME predictions

The information regarding the active site of the proteins was collected from the PDB database. Compound 8 showed the highest binding affinity with the DNA gyrase B of S. aureus with interaction was observed with Gly85, Ile86, Ile51, and Ile175, while for E. coli DNA gyrase B, the compound 20 showed the highest free energy of binding with Asn46, Ile78, Pro79, and Ile90 were present in the binding cavity (Figure 1a and b). Similarly, compound 20 also has relatively higher interaction energy with urate oxide of A. flavus with residues Tyr30, Met32, Cys103, Hs104, and Trp106 in the docked site (Figure 1c). Furthermore, the compounds showed less binding affinity toward the glucosamine-6-phosphate synthase of C. albicans, with relatively higher energy for the docked complex of compound 20, which was observed to have interacted with Leu411, Glu418, Tyr577, and Ala580 (Figure 1d). All the studied compounds demonstrated significant ADME properties, with the number of hydrogen bond donors [1,2], the number of hydrogen bond acceptors [2,3,4,5], and molecular weight of <320, which are considerable (Table 2). None of the studied compounds showed the ability to cross the blood–brain barrier except compound 18, with the nontoxic nature of the selected compounds. Compound 15 was predicted to be soluble, with the rest of the compounds showing moderate solubility in water. All the designed compounds are practicality synthesizable, which can be indicated from the reasonable ranges presented through the ADME calculations.

Figure 1 
                     The docked complex formation (a) between S. aureus DNA gyrase B and compound 8, (b) between E. coli DNA gyrase B and compound 20, (c) between A. flavus urate oxide and compound 20, and (d) between C. albicans glucosamine-6-phosphate synthase and compound 20.
Figure 1

The docked complex formation (a) between S. aureus DNA gyrase B and compound 8, (b) between E. coli DNA gyrase B and compound 20, (c) between A. flavus urate oxide and compound 20, and (d) between C. albicans glucosamine-6-phosphate synthase and compound 20.

Table 2

List ADMET properties calculated for the selected inhibitors

S. no. Compound name QP log P o/w QPP Caco QP log BB QPPMDCK #metab QP log Khsa Percent human oral absorption
1 6 2.42 205.83 −0.422 113.96 4 −0.235 82.535
2 8 1.83 1063.07 −0.402 528.52 3 −0.151 92.02
3 15 3.37 493.71 −0.119 293.40 3 0.085 94.92
4 18 2.92 597.64 0.466 606.98 2 0.301 93.75
5 20 2.40 351.05 −0.366 794.06 3 −0.089 86.60

Predicted 1: octanol/water partition coefficient; 2: Caco-2 cell permeability (nm/s); 3: brain/blood partition coefficient; 4: apparent MDCK cell permeability (nm/s); 5: human serum albumin binding; 6: number of metabolic reactions; 7: percent human oral absorption.

3 Experimental and methodology

3.1 Chemistry

An electro-thermal IA 9100 apparatus was used for measuring melting points. Drying solvents were used for all experiments. Purification of the produced compounds was carried out by crystallization. The IR analyses (KBr disc) were performed using a Pye Unicom Sp-3-300 or a Shimadzu FTIR 8101 PC infrared spectrophotometer. The 1HNMR and 13CNMR spectra were recorded on Varian Mercury VX-300 NMR 500 (75.4) MHz at a spectrophotometer using DMSOd6 as a solvent. TMS was used as an internal reference for all chemical shifts expressed on the δ (ppm) scale. The values of the coupling constant (J) are given in Hz [45,46].

3.1.1 6-Phenyl-3,6-dihydropyridine-2,5-diol (6)

A solution of N-benzyl-2-chloroacetamide 2 (1,500 mg, 8 mmol), diethyl malonate (1.22 mL, 8 mmol), and TEA (1 mL) in butanol (20 mL) was refluxed for 15 h and then concentrated, and the formed precipitate was filtered off, washed with water, dried, and crystallized with a mixture of ethanol + H2O by ratio of 1:3 to obtain white crystals of 6. Yield 1175.1 mg (76%), m.p. 82–84°C. IR (KBr, v, cm−1): 3,274(2O–H) 1,644(C═N), 1,549(C═C). 1H NMR (500 MHz, DMSO-d6) δ 7.42–7.27 (m, 5H), 5.87 (td, J = 5.8, 1.8 Hz, 1H), 5.15 (dt, J = 1.8, 0.9 Hz, 1H), 3.11–2.98 (m, 2H). 13C NMR (125 MHz, chloroform-d) δ 172.57, 158.29, 138.62, 128.43, 127.58, 126.55, 97.57, 59.28, 26.34. Elemental analysis calculated (%) for C11H11NO2 (189): C 69.83; H 5.86; N 7.40. Found: C 69.65; H 5.76; N 7.32.

3.1.2 4-Acetyl-5-methyl-6-phenyl-3,4-dihydropyridin-2(1H)-one (8)

A mixture of N-benzyl-2-chloroacetamide 2 (7,340 mg, 40 mmol), acetyl acetone (4.1 mL, 40 mmol), and TEA (1 mL) was refluxed in butanol (20 mL) for 15 h and then concentrated. The formed precipitate was filtered off, washed with water, dried, and recrystallized with mixture of ethanol + H2O by ratio 1:3 to give brown powder of 8. Yield 6,598 mg (72%), m.p. 80–86°C. IR (KBr, v, cm−1): 3,275(N–H), 1,645(C═O), 1,549(C═C). 1H NMR (500 MHz, DMSO-d6) δ 7.55–7.47 (m, 3H), 7.43–7.36 (m, 2H), 4.70 (tdt, J = 7.7, 2.9, 1.5 Hz, 1H), 2.74 (dd, J = 15.7, 7.8 Hz, 1H), 2.52–2.46 (m, 1H), 2.13 (d, J = 1.5 Hz, 3H), 1.82 (d, J = 1.3 Hz, 3H). 13C NMR (125 MHz, chloroform-d) δ 206.16, 171.14, 137.14, 133.68, 128.84, 128.20, 127.51, 114.39, 52.84, 37.35, 29.09, 16.52.

Elemental analysis calculated (%) for C14H15NO2 (229.11): C 73.34; H 6.59; N 6.11. Found: C 73.29; H 6.46; N 6.6.

3.1.3 6-Phenyl-3,4-dihydropyridine-2,5-dione (12)

A solution of N-benzyl-2-chloroacetamide 2 (1,500 mg, 8 mmol), ethyl cyanoacetate (0.9 mL, 8 m.mol), and TEA (1 mL) in butanol (15 mL) was refluxed for 15 h and then concentrated. The formed precipitate was filtered off, washed with water, dried, and crystallized from butanol to give orange crystals of 12. Yield 1112.8 mg (72%), m.p. 75–78°C. IR (KBr, v, cm−1): 1,641(C═O), 1,529(C═N). 1H NMR (500 MHz, DMSO-d 6) δ 8.19–8.11 (m, 1H), 7.57–7.47 (m, 1H), 3.16–3.09 (m, 1H), 3.02–2.95 (m, 1H). 13C NMR (125 MHz, chloroform-d) δ 193.68, 176.82, 162.36, 135.84, 130.19, 129.74, 129.31, 32.30, 31.61. Elemental analysis calculated (%) for C11H9NO2 (187): C 70.58; H 4.85; N 7.48. Found: C 70.50; H 4.78; N 7.40.

3.1.4 5-Methyl-6-phenyl-3,4-dihydropyridin-2-ol (15)

A solution of N-benzyl-2-chloroacetamide 2 (1,000 mg, 5 mmol), ethyl acetoacetate (0.688 mL, 5 mmol), and TEA (1 mL) in butanol (20 mL) was refluxed for 15 h and then concentrated. The formed precipitate was filtered off, washed with water, dried, and crystallized from butanol to give pale yellow crystals of 15. Yield 795.3 mg (78%), m.p. 79–80°C. IR (KBr, v, cm−1): 3,275(O–H), 1,644(C═N), 1,548(C═C). 1H NMR (500 MHz, DMSO-d 6) δ 7.76–7.70 (m, 1H), 7.55–7.43 (m, 1H), 2.63–2.51 (m, 1H), 1.76 (s, 1H). 13C NMR (125 MHz, chloroform-d) δ 167.90, 147.09, 137.19, 129.73, 128.22, 127.15, 126.12, 30.42, 26.15, 18.90. Elemental analysis calculated (%) for C12H13NO (187.1): C 76.98; H 7.00; N 7.48. Found: 76.90; H 6.94; N 7.40.

3.1.5 4-(Benzyl amino)-1,5-dihydro-2H-imidazole-2-thione (16)

A solution of thiourea (380 mg, 5 mmol), N-benzyl-2-chloroacetamide 2 (1,000 mg, 5 mmol), and sodium (120 mg, 5 mmol) in ethanol (20 mL) was refluxed for 13 h, and then HCl was added dropwise till complete precipitate was obtained. The formed precipitate was filtered off, washed with water, dried, and crystallized from ethanol to give white crystals of 16. Yield 860.5 mg (77%), m.p. 150–152°C. IR (KBr, v, cm−1): 3,270(N–H), 1,641(C═N), 1,311(C═S). 1H NMR (500 MHz, DMSO-d 6) δ 9.40 (t, J = 5.0 Hz, 1H), 8.52 (t, J = 4.5 Hz, 1H), 7.36–7.30 (m, 1H), 7.31 (s, 3H), 7.34–7.22 (m, 2H), 4.52 (dd, J = 5.0, 0.8 Hz, 2H), 4.26 (d, J = 4.6 Hz, 2H). 13C NMR (125 MHz, chloroform-d) δ 188.12, 167.22, 138.11, 128.29, 127.59, 127.49, 47.53, 46.85. Elemental analysis calculated (%) for C10H11N3S (205): C 58.51; H 5.40; N 20.47; S 15.62. Found: C 58.49; H 5.32; N 20.42; S 15.58.

3.1.6 3-Chloro-4,5-diphenyl-1,3-dihydro-2H-pyrrol-2-one (18)

A solution of benzaldehyde (0.54 mL, 5 mmol), N-benzyl-2-chloroacetamide 2 (1,000 mg, 5 mmol), and sodium (120 mg, 5 mmol) in ethanol (20 mL) was refluxed for 17 h, and then HCl was added dropwise till complete precipitate was obtained. The formed precipitate was filtered off, washed with water, dried, and crystallized from ethanol to give yellow crystals of 18. Yield 1,099 mg (75%), m.p. 72–75°C. IR (KBr, v, cm−1): 3,283(N–H), 1,652(C═O), 1,532(C═C). 1H NMR (500 MHz, DMSO-d 6) δ 9.28 (s, 1H), 7.60–7.54 (m, 2H), 7.53–7.46 (m, 1H), 7.44–7.34 (m, 4H), 7.34–7.26 (m, 3H), 6.14 (s, 1H). 13C NMR (125 MHz, chloroform-d) δ 169.86, 135.75, 131.13, 130.22, 128.92, 128.58, 128.10, 128.00, 127.62, 126.32, 108.92, 57.22. Elemental analysis calculated (%) for C16H12ClNO (269): C 71.25; H 4.48; Cl 13.14; N 5.19. Found: C 71.20; H 4.42; Cl 13.08; N 5.07.

3.1.7 4-Benzoyl-6-phenyl-5-thioxopiperazin-2-one (20)

A solution of benzoyl isothiocyanate (40 mmol), N-benzyl-2-chloroacetamide 2 (7,340 mg, 40 mmol) in dioxane (23 mL) was refluxed for 17 h and then poured into crushed ice. The formed precipitate was filtered off, washed with water, dried, and crystallized from dioxane to give pale orange crystals of 20. Yield 8,680 mg (70%), m.p. 80–81°C. IR (KBr, v, cm−1): 3,272(N–H), 1,638(C═O), 1,552(C═C), 1,226(C═S). 1H NMR (500 MHz, DMSO-d 6) δ 7.88–7.82 (m, 2H), 7.58–7.46 (m, 3H), 7.45–7.38 (m, 2H), 7.38–7.27 (m, 3H), 7.20 (d, J = 7.5 Hz, 1H), 5.64 (dd, J = 7.6, 0.7 Hz, 1H), 4.81–4.70 (m, 2H). 13C NMR (125 MHz, chloroform-d) δ 200.42, 169.86, 166.14, 138.09, 135.23, 132.07, 129.12, 128.71, 128.04, 127.98, 62.79, 49.38. Elemental analysis calculated (%) for C17H14N2O2S (310): C 65.79; H 4.55; N 9.03; S 10.33. Found: C 65.72; H 4.50; N 8.99; S 10.28.

3.2 Biology

A panel of Gram-positive bacteria (Staphylococcus aureus) and Gram-negative bacteria were examined for the antimicrobial action of the synthesized samples (Escherichia coli). The antifungal activities of the compounds were tested against two fungi (Candida albicans and Aspergillus flavus). Each sample was dissolved in DMSO, and 1 mg/mL of solutions were prepared separately as paper discs of Whitman filter paper of the standard size (5 cm) and were cut and sterilized in an autoclave. Petri dishes with nutrient agar media (agar 20 g + beef extract 3 g + peptone 5 g) were seeded with Staphylococcus aureus, E. coli, Candida albicans, and Aspergillus flavus, and the paper discs soaked in the desired concentration of the complex solution were placed aseptically. The Petri dishes were incubated at 36°C, and the inhibition zones were recorded after 24 h of incubation. Each test was repeated three times at the same solvents and solution concentration, and the antibacterial activities of a common standard antibiotic ampicillin and antifungal colitrimazole were recorded by using the same procedure as mentioned earlier. The percent activity index for the complex was calculated by the following formula:

% Activity index =   Zone of inhibition by test compound (diametre) Zone of inhibition by standard  ( diametre ) × 100 .

3.3 Computational studies

3.3.1 Molecular docking and ADME analyses

As per the reference of the standard compounds used for the evaluation of the activities of the designed compounds, the protein target for each organism was selected from the Protein Data Bank (PDB) listed in Table 3. To study the effect of the drugs on bacterial protein, the DNA gyrase B (PDB IDs – 3U2K, 1KZN) was selected, but for A. flavus, it was urate oxide (PDB ID – 1R51), and for C. albicans, the glucosamine-6-phosphate synthase (PDB ID – 2PUV) was used. The selected structures were optimized using the utilities JAGUAR modules [42] present in MAESTRO (Schrödinger Release 2018-1: Maestro, Schrödinger, LLC, New York, NY, 2018). Consequently, molecular docking was performed between the designed molecules and target proteins using Autodock vina [43]. The best-docked conformations were statistically characterized through the combination of the free energy functional, the Lamarckian Genetic Algorithm, and the empirical force field [2]. The space of grid dimensional was set for 40 × 40 × 40 Å along the XYZ directions with varied central coordinates, and the maximum efficiency range was used in the parameters for the optimum results.

Table 3

List of molecular docking-based generated free energies of binding for the designed inhibitors

S. no. Compound Free energy of binding (kcal/mol)
S. aureus E. coli A. flavus C. albicans Glucosamine-6-phosphate synthase
DNA gyrase B DNA gyrase B Urate oxide
1. 6 −6.9 −6.6 −6.7 −6.0
2. 8 −8.2 −7.5 −7.7 −6.6
3. 15 −7.7 −7.2 −6.9 −5.9
4. 18 −7.5 −6.8 −7.4 −6.6
5. 20 −7.7 7.6 8.2 6.9

Furthermore, the ADME properties for each compound were calculated using QikProp tools present in the Schrodinger 2020-2. ADME (absorption, distribution, metabolism, and excretion), including drug-likeness analysis, is essential in the drug discovery, which accommodates to make a reasonable decisiveness on whether inhibitors can be conducted to a biological system [40,41]. A potent antagonistic interaction of inhibitors with a receptor protein or enzyme cannot guarantee the ability of an inhibitor as a drug; therefore, ADME assessment is essential in the drug development. Inhibitors with lower ADME properties and high toxicity effects on the biological systems are often the dominant explanation of most failed medicines in the clinical phase of experiments. From the output of some ADME and drug-likeness properties presented in Table 2, it was observed that most compounds have one or two violations of Lipinski’s rule.

4 Conclusion

α-Chloroacetyl derivative 2 underwent alkylation followed by intramolecular cyclization using carbonyl to provide pyridones. Target 2 underwent heterocyclization with benzaldehyde and benzoyl isothiocyanate to yield derivatives of 2-pyridone. The synthesized compound was tested for its antimicrobial activity against the Gram-positive (Staphylococcus aureus) and the Gram-negative (Escherichia coli) bacteria. The antifungal activities of the compounds were tested against two fungi (Candida albicans and Aspergillus flavus). Compound 20 has the highest antibacterial activity against Gram-positive (Staphylococcus aureus) and Gram-negative (Escherichia coli) bacteria and antifungal activity against Candida albicans. Such compounds that had antimicrobial activity should be further reused for the inhibitory microbial growth. The docking results show that compound 2 had the best docking free energy results toward S. aureus, while compound 20 was the effective compound toward the selected bacterial and fungal proteins. These results agreed with the experimental results.

Acknowledgements

The authors are very thankful to all the associated personnel in any reference that contributed to/for this research. M.H.A. is especially thankful to Taif University Researcher’s Supporting Project Number (TURSP-2020/91), Taif University, Taif, Saudi Arabia.

  1. Funding information: The authors state no funding involved.

  2. Conflict of interest: The authors state no conflict of interest.

  3. Data availability statement: The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.

References

[1] Yuan C, Zhang H, Yuan M, Xie L, Cao X. Synthesis of 1, 4-diazepinone derivatives via a domino aza-Michael/SN 2 cyclization of 1-azadienes with α-halogenoacetamides. Org Biomol Chem. 2020;18(6):1082–6.10.1039/C9OB02626FSearch in Google Scholar PubMed

[2] Jin Q, Zhang D, Zhang J. Facile synthesis of 1, 2, 4, 5-tetrahydro-1, 4-benzodiazepin-3-ones via cyclization of N-alkoxy α-halogenoacetamides with N-(2-chloromethyl) aryl amides. Org Biomol Chem. 2019;17(45):9708–11.10.1039/C9OB02260KSearch in Google Scholar

[3] Fantinati A, Zanirato V, Marchetti P, Trapella C. The fascinating chemistry of α-haloamides. ChemistryOpen. 2020;9:100–70.10.1002/open.201900220Search in Google Scholar PubMed PubMed Central

[4] Zhou C, Ni J, Zhang D, Sun C. Cellulosic adsorbent functionalized with macrocyclic pyridone pentamer for selectively removing metal cations from aqueous solutions. Carbohydr Polym. 2019;217:1–5.10.1016/j.carbpol.2019.04.048Search in Google Scholar PubMed

[5] Christoforow A, Wilke J, Binici A, Pahl A, Ostermann C, Sievers S, et al. Design, synthesis, and phenotypic profiling of pyrano-furo-pyridone pseudo natural products. Angew Chem Int Ed. 2019;58(41):14715–23.10.1002/anie.201907853Search in Google Scholar PubMed PubMed Central

[6] Kulangiappar K, Anbukulandainathan M, Raju T. Synthetic communications: an international journal for rapid communication of synthetic organic chemistry. Synth Commun. 2014;1(44):2494–502.10.1080/00397911.2014.905599Search in Google Scholar

[7] Szyc Ł, Guo J, Yang M, Dreyer J, Tolstoy PM, Nibbering ET, et al. The hydrogen-bonded 2-pyridone dimer model system. 1. Combined NMR and FT-IR spectroscopy study. J Phys Chem A. 2010;114(29):7749–60.10.1021/jp103630wSearch in Google Scholar PubMed

[8] Xie W, Wu Y, Zhang J, Mei Q, Zhang Y, Zhu N, et al. design, synthesis and biological evaluations of novel pyridone-thiazole hybrid molecules as antitumor agents. Eur J Med Chem. 2018;145:35–40.10.1016/j.ejmech.2017.12.038Search in Google Scholar PubMed

[9] Li L-N, Wang L, Cheng YN, Cao ZQ, Zhang XK, Guo XL. Discovery and characterization of 4-hydroxy-2-pyridone derivative sambutoxin as a potent and promising anticancer drug candidate: activity and molecular mechanism. Mol Pharmaceutics. 2018;15(11):4898–911.10.1021/acs.molpharmaceut.8b00525Search in Google Scholar PubMed

[10] Bai H, Sun R, Chen X, Yang L, Huang C. Microwave-assisted, solvent-free, three-component domino protocol: efficient synthesis of polysubstituted-2-pyridone derivatives. ChemistrySelect. 2018;3(17):4635–8.10.1002/slct.201800606Search in Google Scholar

[11] Gonçalves DS, de S Melo SM, Jacomini AP, J V da Silva M, Pianoski KE, Ames FQ, et al. Synthesis of novel 3, 5, 6-trisubstituted 2-pyridone derivatives and evaluation for their anti-inflammatory activity. Bioorg Med Chem. 2020;28(12):115549.10.1016/j.bmc.2020.115549Search in Google Scholar PubMed

[12] Mahmoud MR, Abu El-Azm FSM, Ali AT, Ali YM. Synthesis and antimicrobial evaluation of some novel dithiolane, thiophene, coumarin, and 2-pyridone derivatives. Synth Commun. 2017;47(17):1591–600.10.1080/00397911.2017.1336776Search in Google Scholar

[13] Pandit AB, Savant MM, Ladva KD. An efficient one-pot synthesis of highly substituted pyridone derivatives and their antimicrobial and antifungal activity. J Heterocycl Chem. 2018;55(4):983–7.10.1002/jhet.3128Search in Google Scholar

[14] Ibrahim M. One-pot synthesis, characterization and antimicrobial activity of new 3-cyano-4-alkyl-6-(2, 5-dichlorothiophen-3-yl)-2 (1H)-pyridones. Jordan J Chem (JJC). 2015;10(2):98–107.10.12816/0026450Search in Google Scholar

[15] Gaffer H, Elapasery M, Abbas D, Allam E. Synthesis of some new aryl-azo derivatives clubbed with pyridone and evaluating their biological broadcast. Egypt J Chem. 2020;63(3):1087–99.10.21608/ejchem.2020.22449.2335Search in Google Scholar

[16] Maeng CY et al. Pharmaceutical composition comprising pyridone derivatives. Google Pat; 2018.Search in Google Scholar

[17] Azzam RA, Elboshi HA, Elgemeie GH. Novel synthesis and antiviral evaluation of new benzothiazole-bearing N-sulfonamide 2-pyridone derivatives as USP7 enzyme inhibitors. ACS Omega. 2020;5(46):30023–36.10.1021/acsomega.0c04424Search in Google Scholar PubMed PubMed Central

[18] Biswas A, Maity S, Pan S, Samanta R. Transition metal-catalysed direct C–H bond functionalizations of 2-pyridone beyond C3-selectivity. Chemistry – Asian J. 2020;15(14):2092–109.10.1002/asia.202000506Search in Google Scholar PubMed

[19] Chand K, Alsoghier HM, Chaves S, Santos MA. Tacrine-(hydroxybenzoyl-pyridone) hybrids as potential multifunctional anti-Alzheimer’s agents: AChE inhibition, antioxidant activity and metal chelating capacity. J Inorg Biochem. 2016;163:266–77.10.1016/j.jinorgbio.2016.05.005Search in Google Scholar PubMed

[20] Sun C, Huang Z, Wang J, Rao L, Zhang J, Yu J, et al. Modification of microcrystalline cellulose with pyridone derivatives for removal of cationic dyes from aqueous solutions. Cellulose. 2016;23(5):2917–27.10.1007/s10570-016-1024-9Search in Google Scholar

[21] Elapasery M, Shakra S, Abbas D, Gaffer HE, Allam EA. Synthesis of some azo disperse dyes based on pyridone moiety and their application on polyester fabrics. Egyptian J Chem. 2017;60(Conference Issue (The 8th International Conference of The Textile Research Division (ICTRD 2017), National Research Centre, Cairo 12622, Egypt.):97–102.Search in Google Scholar

[22] Mijin D, et al. synthesis, solvatochromism, and biological activity of novel azo dyes bearing 2-pyridone and benzimidazole moieties. Turkish J Chem. 2018;42(3):896–907.Search in Google Scholar

[23] Zhao X-L, Qian H-F, Huang W. Construction of benzothiazole/pyridone based bi-heterocyclic dyes and their NiII and CuII complexes. Dye Pigment. 2018;149:796–803.10.1016/j.dyepig.2017.11.054Search in Google Scholar

[24] Al-Etaibi AM, El-Apasery MA. A comprehensive review on the synthesis and versatile applications of biologically active pyridone-based disperse dyes. Int J Environ Res Public Health. 2020;17(13):4714.10.3390/ijerph17134714Search in Google Scholar PubMed PubMed Central

[25] Kadam ML, D, Patil, Sekar N. Fluorescent carbazole based pyridone dyes–synthesis, solvatochromism, linear and nonlinear optical properties. Opt Mater. 2018;85:308–18.10.1016/j.optmat.2018.08.072Search in Google Scholar

[26] Jessen HJ, Gademann K. 4-Hydroxy-2-pyridone alkaloids: structures and synthetic approaches. Nat product Rep. 2010;27(8):1168–85.10.1039/b911516cSearch in Google Scholar PubMed

[27] Jessen HJ, Schumacher A, Schmid F, Pfaltz A, Gademann K. Catalytic enantioselective total synthesis of (+)-torrubiellone C. Org Lett. 2011;13(16):4368–70.10.1021/ol201692hSearch in Google Scholar PubMed

[28] Fischer CB, Steininger H, Stephenson DS, Zipse H. Catalysis of aminolysis of p-nitrophenyl acetate by 2-pyridones. J Phys Org Chem. 2005;18(9):901–7.10.1002/poc.914Search in Google Scholar

[29] Wang LH, Zipse H. Bifunctional catalysis of ester aminolysis–a computational and experimental study. Liebigs Annalen. 1996;1996(10):1501–9.10.1002/jlac.199619961003Search in Google Scholar

[30] Fischer CB, Polborn K, Steininger H, Zipse H. Synthesis and solid-state structures of alkyl-substituted 3-cyano-2-pyridones. Z Naturforschung B. 2004;59(10):1121–31.10.1515/znb-2004-1008Search in Google Scholar

[31] Donets PA, Cramer N. Ligand-controlled regiodivergent nickel-catalyzed annulation of pyridones. Angew Chem. 2015;127(2):643–7.10.1002/ange.201409669Search in Google Scholar

[32] Wang P, Verma P, Xia G, Shi J, Qiao JX, Tao S, et al. Ligand-accelerated non-directed C–H functionalization of arenes. Nature. 2017;551(7681):489–93.10.1038/nature24632Search in Google Scholar PubMed PubMed Central

[33] Poomathi N, Perumal P, Ramakrishna S. An efficient and eco-friendly synthesis of 2-pyridones and functionalized azaxanthone frameworks via indium triflate catalyzed domino reaction. Green Chem. 2017;19(11):2524–9.10.1039/C6GC03440CSearch in Google Scholar

[34] Torres M, Gil S, Parra M. New synthetic methods to 2-pyridone rings. Curr Org Chem. 2005;9(17):1757–79.10.2174/138527205774610886Search in Google Scholar

[35] Heravi MM, Hamidi H. Recent advances in synthesis of 2-pyridones: a key heterocycle is revisited. J Iran Chem Soc. 2013;10(2):265–73.10.1007/s13738-012-0155-7Search in Google Scholar

[36] Samzadeh-Kermani A. Heteropolyacid-catalyzed one-pot synthesis of 2-pyridone derivatives. Synlett. 2016;27(03):461–4.10.1055/s-0035-1560824Search in Google Scholar

[37] Babaee S, Zarei M, Sepehrmansourie H, Zolfigol MA, Rostamnia S. Synthesis of metal–organic frameworks MIL-101 (Cr)-NH2 containing phosphorous acid functional groups: Application for the synthesis of N-Amino-2-pyridone and pyrano [2, 3-c] pyrazole derivatives via a cooperative vinylogous anomeric-based oxidation. ACS Omega. 2020;5(12):6240–9.10.1021/acsomega.9b02133Search in Google Scholar PubMed PubMed Central

[38] Jia H, Song Y, Yu J, Zhan P, Rai D, Liang X, et al. design, synthesis and primary biological evaluation of the novel 2-pyridone derivatives as potent non-nucleoside HBV inhibitors. Eur J Med Chem. 2017;136:144–53.10.1016/j.ejmech.2017.04.048Search in Google Scholar PubMed

[39] Abdellattif MH, Abdel-Rahman A, Arief M, Mouneir SM, Ali A, Hussien MA, et al. Novel 2-Hydroselenonicotinonitriles and Selenopheno[2, 3-b]pyridines: Efficient Synthesis, Molecular Docking-DFT Modeling, and Antimicrobial Assessment. Front Chem. May 2021;9:10. 10.3389/fchem.2021.672503.Search in Google Scholar PubMed PubMed Central

[40] Rai SK, Khanam S, Khanna RS, Tewari AK. Design and synthesis of 2-pyridone based flexible dimers and their conformational study through X-ray diffraction and density functional theory: perspective of cyclooxygenase-2 inhibition. Cryst Growth Des. 2015;15(3):1430–9.10.1021/cg501793vSearch in Google Scholar

[41] Amer MMK, Aziz MA, Shehab WS, Abdellattif MH, Mouneir SM. Recent advances in chemistry and pharmacological aspects of 2-pyridone scaffolds. J Saudichemical Soc. 2021;25(6):101259. 10.1016/ j.jscs.2021.101259.Search in Google Scholar

[42] Abdellattif MH, Shahbaaz M, Arief MMH, Hussien MA. Oxazinethione derivatives as a precursor to pyrazolone and pyrimidine derivatives: Synthesis, biological activities, molecular modeling, ADME, and molecular dynamics studies. Molecules. 2021;26:5482. 10.3390/molecules26185482.Search in Google Scholar PubMed PubMed Central

[43] Ochevarov AD, Harder E, Hughes TF, Greenwood JR, Braden DA, Philipp DM, et al. A high-performance quantum chemistry software program with strengths in life and materials sciences. Int J Quant Chem. 2013;113:2110–42. 10.1002/qua.24481.Search in Google Scholar

[44] Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization and multithreading. J Comput Chem. 2010;31:455–61. 10.1002/jcc.21334.Search in Google Scholar PubMed PubMed Central

[45] Zhou L, Dai S, Xu S, She Y, Li Y, Leveneur S, et al. Piezoelectric effect synergistically enhances the performance of Ti32-oxo cluster/BaTiO3/CuS p-n heterojunction photocatalytic degradation of pollutants. Ind Chem React Eng. 2021;291:120019. 10.1016/j.apcatb.2021.120019. Search in Google Scholar

[46] Xu S, Zhu Q, Xu S, Yuan M, Lin X, Lin W, et al. The phase behavior of n-ethylpyridinium tetrafluoroborate and sodium-based salts ATPS and its application in 2-chlorophenol extraction. Chin J Chem Eng. May 2021;33:76–82. 10.1016/j.cjche.2020.07.024.Search in Google Scholar

Received: 2021-08-01
Revised: 2021-09-23
Accepted: 2021-10-19
Published Online: 2022-03-23

© 2022 Eman O. Hamed et al., published by De Gruyter

This work is licensed under the Creative Commons Attribution 4.0 International License.

Downloaded on 5.5.2024 from https://www.degruyter.com/document/doi/10.1515/hc-2022-0004/html
Scroll to top button