Next Article in Journal
Development of HPLC Method for Simultaneous Determination of Ibuprofen and Chlorpheniramine Maleate
Next Article in Special Issue
Effect of Aqueous Extracts of Quercus resinosa on the Mechanical Behavior of Bigels
Previous Article in Journal / Special Issue
Melt Fusion Techniques for Solubility Enhancement: A Comparison of Hot Melt Extrusion and KinetiSol® Technologies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

New Quinazolin-4(3H)-one Derivatives Incorporating Hydrazone and Pyrazole Scaffolds as Antimicrobial Agents Targeting DNA Gyraze Enzyme

1
Department of Therapeutic Chemistry, National Research Centre, Dokki, Cairo 12622, Egypt
2
Department of Pharmaceutical Medicinal Chemistry, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11754, Egypt
3
Department of Microbial Chemistry, National Research Centre, Dokki, Cairo 12622, Egypt
*
Author to whom correspondence should be addressed.
Sci. Pharm. 2022, 90(3), 52; https://doi.org/10.3390/scipharm90030052
Submission received: 2 August 2022 / Revised: 14 August 2022 / Accepted: 17 August 2022 / Published: 26 August 2022
(This article belongs to the Special Issue Feature Papers in Scientia Pharmaceutica)

Abstract

:
The present work includes the synthesis of a new series of quinazolin-4(3H)-one compounds (4af, 5ad) as antimicrobial agents. The starting compound, 2-hydrazinylquinazolin-4(3H)-one (2), was synthesized and treated with different carbonyl compounds to afford the hydrazone derivatives 4af. In addition, the hydrazone derivatives 4ad were treated with a DMF/POCl3 mixture to give the formyl-pyrazole derivatives 5ad. All the target compounds were evaluated as antimicrobial agents against four bacterial and four fungal strains. The majority of the tested compounds showed potent antimicrobial activity compared with the reference antibiotics. The most potent antimicrobial activity was shown by 5a with MIC values in the range (1–16) μg/mL. In addition, the most potent compounds against E. coli were evaluated for their inhibitory activity against E. coli DNA gyrase, whereas the target compounds 4a, 5a, 5c, and 5d showed the most potent inhibition to the target enzyme with IC50 values ranging from 3.19 to 4.17 µM. Furthermore, molecular docking studies were performed for the most active compounds against the target E. coli DNA gyrase to determine their binding affinity within the enzyme’s active site. Moreover, ADME evaluations of these compounds predicted their high oral bioavailability and good GI absorption.

1. Introduction

In the last few decades, the risk of infectious diseases has emerged as a growing threat to global health [1], where severe infections of several resistant bacterial strains have become very common, which have shown a diverse resistance pattern to many commonly used antibiotics [2,3]. Thus, the risk of antimicrobial resistance has led to the pressing need to discover new antimicrobial molecules having structural features different from those of the present antibiotics to defeat the resistance mechanisms [4]. Most clinically significant antibiotics such as β-lactams, quinolones, carbapenems, and aminoglycosides have a specific mode of action as microbial enzyme inhibitors; they target specifically the enzymes that are responsible of vital functions such as the biosynthesis of the cell wall, proteins, nucleic acids, and metabolites [5,6]. However after decades of using these antibiotics, their binding to the target enzymes has been obstructed by different resistance mechanisms [7]. Among these enzymes, DNA gyrase, which is present in all microbial cells, plays a pivotal role in bacterial cell cycle progression by introducing negative supercoils into DNA during replication [8,9]. As a result, DNA gyrase inhibition leads to cell death and prevents resistance development [10].
On the other hand, quinazolin-4(3H)-one compounds have attracted great interest due to their significant pharmacological activities, including anticancer [11,12,13], antimicrobial [14,15,16], anti-inflammatory [17,18], analgesic [19,20], antimalarial [21,22], and antiviral [23] activities. Moreover, several quinazolinone derivatives have been reported as potential antimicrobial agents and DNA gyrase inhibitors I–III (Figure 1) [24,25,26].
In addition, hydrazones and pyrazole derivatives have received renewed interest due to their various important biological activities [27,28,29,30], and some recent studies have explored novel compounds carrying hydrazone and pyrazole moieties as promising candidates to be potent DNA gyrase inhibitors IV–VII (Figure 1) [31,32,33,34].
Molecular hybridization is an important concept in drug design; it is based on the combination of different pharmacophores to produce innovative hybrid compounds with improved bioactive efficacy and is considered as one of the recent strategies used for developing new antimicrobial agents to overcome resistance mechanisms [35].
In view of the above topics, the present work includes the design and synthesis of novel hybrid compounds having a quinazolin-4(3H)-one nucleus combined with hydrazone or 4-formylpyrazole moieties (4a4f, 5a5d) to obtain potent antimicrobial agents targeting the DNA gyrase enzyme. The in vitro antimicrobial activities of the target compounds were evaluated against four bacterial and four fungal strains. Molecular docking investigations were also performed for the target compounds against E. coli DNA gyrase to determine the binding affinity of the compounds and their binding style within the enzyme’s active site.

2. Materials and Methods

2.1. Chemistry

2.1.1. General Information

All melting points were uncorrected and were taken in open capillary tubes using an Electro thermal IA9100 digital melting point apparatus. Elemental microanalyses were carried out at the Micro Analytical Unit at Cairo University. The ESI-mass spectra were measured using an Advion Compact Mass Spectrometer (CMS) NY (New York), USA. Infrared spectra were recorded at the National Research Centre, by using the KBr disc technique on a Jasco FT/IR-360 plus Infrared spectrometer in the range (400–4000 cm−1), made in Japan. 1H NMR and 13C NMR spectra were recorded on a JEOL (Tokyo, Japan) High Performance Digital FT-NMR S (500/125 MHz) in the presence of TMS as the internal standard. The follow-up of the reactions and checking the purity of the compounds were performed by TLC on silica-gel-precoated aluminum sheets (Type 60, F 254, Merck, Darmstadt, Germany) using chloroform/methanol (3:1, v/v), and the spots were detected by exposure to a UV lamp at δ 254 nanometers for a few seconds and by iodine vapor. The chemical names given for the synthesized compounds were according to the IUPAC system. The starting compound 2-thioxo-2,3-dihydroquinazolin-4(1H)-one was prepared by the reported method [36].

2.1.2. Synthesis of 2-hydrazinylquinazolin-4(3H)-one (2)

A mixture of compound 1 (8.91 g, 0.05 mol) and hydrazine hydrate 100% (4 mL) in absolute ethanol (50 mL) was refluxed for 12 h. The excess solvent was evaporated till dryness under a vacuum. Then, the obtained solid was treated with cold water, collected by filtration, and recrystallized from ethanol to give compound 2.
Yield 72%, white powder, m.p. 361–362 °C (Lit. m.p. 355 °C [37]); IR (KBr, υmax/cm−1): 3431, 3308, 3187 (NH), 3036 (CH-aromatic), 1674 (C=O), 1601 (C=N). 1H NMR (500 MHz, DMSO-d6, δ ppm): 7.13 (d, 1H, J = 7.6 Hz, Ar-H), 7.42 (d, 1H, J = 8.6 Hz, Ar-H), 7.63–7.81 (m, 2H, Ar-H), 8.09 (s, 2H, NH2, D2O exchangeable), 8.12, 10.24 (2 s, 2H, 2NH, D2O exchangeable). 13C NMR (125 MHz, DMSO-d6, δ ppm): 122.95, 126.83, 127.02, 127.38, 134.52, 146.44, 154.83 (Ar-C, C=N), 163.91(C=O). Anal. Calcd. for C8H8N4O (176.18): C, 54.54; H, 4.58; N, 31.80%; Found: C, 54.78; H, 4.86; N, 31.55%. ESI-MS: m/z = 175.13 [M-H+].

2.1.3. Synthesis of 2-(2-ethylidene-hydrazinyl)quinazolin-4(3H)-one Derivatives 4a–f

A mixture of the 2-hydrazinyl derivative 2 (0.88 g, 5 mmol) and the appropriate carbonyl compound 3af (5 mmol) in glacial acetic acid (20 mL) was refluxed for 6 h. The mixture was poured onto an ice water mixture, and the obtained solid was collected by filtration and recrystallized from acetone to give the corresponding target compound 4af.

2-(2-(1-(Furan-2-yl)ethylidene)hydrazinyl)quinazolin-4(3H)-one (4a)

Yield 74%, light beige powder, m.p. 220–221 °C. IR (KBr, υmax/cm−1): 3420, 3114 (NH), 3041 (CH-aromatic), 2919, 2851 (CH-aliphatic), 1683 (C=O), 1618, 1579 (C=N). 1H NMR (500 MHz, DMSO-d6, δ ppm): 2.24 (s, 3H, N=C–CH3), 6.56 (d, 1H, J = 8.6 Hz, Ar-H), 7.09–7.21 (m, 2H, Ar-H), 7.42 (d, 1H, J = 8.6 Hz, Ar-H), 7.59–7.70 (m, 2H, Ar-H), 7.84 (d, 1H, J = 7.6 Hz, Ar-H), 10.35, 11.18 (2 s, 2H, 2NH, D2O exchangeable). 13C NMR (125 MHz, DMSO-d6, δ ppm): 16.90 (CH3), 110.21, 110.66, 122.89, 126.93, 127.49, 127.74, 134.42, 139.98, 140.45, 146.73, 155.02, 156.79 (Ar-C, 2C=N), 162.18 (C=O). Anal. Calcd. for C14H12N4O2 (268.28): C, 62.68; H, 4.51; N, 20.88%; Found: C, 62.94; H, 4.25; N, 21.09%. ESI–MS: m/z = 267.31 [M-H+].

2-(2-(1-(Thiophen-2-yl)ethylidene)hydrazinyl)quinazolin-4(3H)-one (4b)

Yield 71%, brown powder, m.p. 278 °C. IR (KBr, υmax/cm−1): 3452, 3164 (NH), 3044 (CH-aromatic), 2919, 2858 (CH-aliphatic), 1680 (C=O), 1630, 1582 (C=N). 1H NMR (500 MHz, DMSO-d6, δ ppm): 2.36 (s, 3H, N=C–CH3), 7.06 (d, 1H, J = 7.2 Hz, Ar-H), 7.14 (d, 1H, J = 7.5 Hz, Ar-H), 7.42–7.51 (m, 2H, Ar-H), 7.67–7.72 (m, 2H, Ar-H), 7.85 (d, 1H, J = 7.7 Hz, Ar-H), 10.29, 11.09 (2 s, 2H, 2NH, D2O exchangeable). 13C NMR (125 MHz, DMSO-d6, δ ppm): 15.12 (CH3), 121.11, 122.68, 124.55, 125.03, 127.08, 127.40, 134.38, 140.03, 141.01, 146.87, 155.18, 156.71 (Ar-C, 2C=N), 161.90 (C=O). Anal. Calcd. for C14H12N4OS (284.34): C, 59.14; H, 4.25; N, 19.70; S, 11.28%; Found: C, 59.42; H, 4.58; N, 19.98; S, 10.97%. ESI–MS: m/z = 283.27 [M-H+].

2-(2-(1-(Pyridin-2-yl)ethylidene)hydrazinyl)quinazolin-4(3H)-one (4c)

Yield 78%, pale yellow powder, m.p. 213–214 °C. IR (KBr, υmax/cm−1): 3432, 3158 (NH), 3063 (CH-aromatic), 2919, 2852 (CH-aliphatic), 1689 (C=O), 1622, 1589 (C=N). 1H NMR (500 MHz, DMSO-d6, δ ppm): 2.40 (s, 3H, N=C–CH3), 7.12 (d, 1H, J = 7.6 Hz, Ar-H), 7.27–7.32 (m, 2H, Ar-H), 7.48–7.59 (m, 2H, Ar-H), 7.76–7.87 (m, 2H, Ar-H), 8.53 (d, 1H, J = 6.5 Hz, Ar-H), 10.46, 11.11 (2 s, 2H, 2NH, D2O exchangeable).13C NMR (125 MHz, DMSO-d6, δ ppm): 13.16 (CH3), 122.76, 123.97, 124.92, 126.91, 127.35, 127.85, 135.77, 136.55, 145.12, 148.79, 149.97, 153.73, 155.95 (Ar-C, 2C=N), 161.59 (C=O). Anal. Calcd. for C15H13N5O (279.30): C, 64.51; H, 4.69; N, 25.07%; Found: C, 64.79; H, 4.85; N, 24.88%. ESI–MS: m/z = 278.27 [M-H+].

2-(2-(1-(4-Methoxyphenyl)ethylidene)hydrazinyl)quinazolin-4(3H)-one (4d)

Yield 69%, greenish yellow powder, m.p. 287–288 °C. IR (KBr, υmax/cm−1): 3425, 3191 (NH), 3056 (CH-aromatic), 2919, 2849 (CH-aliphatic), 1694 (C=O), 1638, 1591 (C=N). 1H NMR (500 MHz, DMSO-d6, δ ppm): 2.33 (s, 3H, N=C–CH3), 3.77 (s, 3H, OCH3), 7.14 (d, 1H, J = 7.6 Hz, Ar-H), 7.30 (d, 2H, J = 8.2 Hz, Ar-H), 7.44 (d, 1H, J = 8.6 Hz, Ar-H), 7.59–7.62 (m, 1H, Ar-H), 7.83 (d, 1H, J = 7.6 Hz, Ar-H), 8.11 (d, 2H, J = 8.2 Hz, Ar-H), 10.34, 11.22 (2 s, 2H, 2NH, D2O exchangeable). 13C NMR (125 MHz, DMSO-d6, δ ppm): 16.21 (CH3), 55.78 (OCH3), 114.54, 122.89, 126.87, 127.22, 127.56, 127.99, 129.23, 134.54, 146.90, 147.76, 155.01, 160.44 (Ar-C, 2C=N), 161.97 (C=O). Anal. Calcd. for C17H16N4O2 (308.34): C, 66.22; H, 5.23; N, 18.17%; Found: C, 66.43; H, 5.41; N, 17.92%. ESI–MS: m/z = 307.39 [M-H+].

2-(2-(4-Methylcyclohexylidene)hydrazinyl)quinazolin-4(3H)-one (4e)

Yield 76%, orange powder, m.p. 133 °C. IR (KBr, υmax/cm−1): 3422, 3192 (2NH), 3036 (CH-aromatic), 2923, 2861 (CH-aliphatic), 1676 (C=O), 1638, 1598 (C=N). 1H NMR (500 MHz, DMSO-d6, δ ppm): 0.88 (d, 3H, J = 6.7 Hz, CH-CH3), 1.21–1.25 (m, 1H, CH-CH3), 1.40–2.05 (m, 8H, 4CH2), 7.12 (d, 1H, J = 7.6 Hz, Ar-H), 7.42 (d, 1H, J = 8.6 Hz, Ar-H), 7.62 (d, 1H, J = 8.95 Hz, Ar-H), 7.82 (d, 1H, J = 8.5 Hz, Ar-H), 10.45, 11.21 (2 s, 2H, 2NH, D2O exchangeable). 13C NMR (125 MHz, DMSO-d6, δ ppm): 21.75 (CH3), 26.78, 31.21, 32.10, 35.55 (4CH2, CH), 122.84, 126.87, 127.03, 127.39, 134.53, 146.60 (Ar-C) 154.45, 161.28, 162.77 (2 C=N, C=O). Anal. Calcd. for C15H18N4O (270.34): C, 66.64; H, 6.71; N 20.73%; Found: C, 66.47; H, 6.44; N, 20.49%. ESI−MS: m/z = 269.48 [M-H+].

2-(2-(2-Oxoindolin-3-ylidene)hydrazinyl)quinazolin-4(3H)-one (4f)

Yield 79%, yellow powder, m.p. 304 °C. IR (KBr, υmax/cm−1): 3432, 3201 (NH), 3062 (CH-aromatic), 1699, 1671 (C=O), 1633, 1596 (C=N). 1H NMR (500 MHz, DMSO-d6, δ ppm): 7.09 (d, 1H, J = 7.6 Hz, Ar-H), 7.29–7.36 (m, 1H, Ar-H), 7.40–7.48 (m, 2H, Ar-H), 7.54–7.81 (m, 3H, Ar-H), 7.91(d, 1H, J = 7.6 Hz, Ar-H), 10.50, 10.91, 11.22 (3 s, 3H, 3NH, D2O exchangeable). 13C NMR (125 MHz, DMSO-d6, δ ppm): 118.02, 119.35, 122.95, 124.72, 126.83, 127.12, 127.36, 128.61, 130.35, 134.52, 136.64, 140.95, 146.36, 154.73 (Ar-C, 2C=N), 161.97, 169.03 (2 C=O) Anal. Calcd. for C16H11N5O2 (305.30): C, 62.95; H, 3.63; N, 22.94%; Found: C, 62.61; H, 3.39; N, 22.66%. ESI−MS: m/z = 304.24 [M-H+].

2.1.4. Synthesis of 1-(4-oxo-3,4-dihydroquinazolin-2-yl)-1H-pyrazole-4-carbaldehyde Derivatives (5ad)

To a cold solution of phosphorus oxychloride (1.2 mL) in N,N-dimethyl formamide DMF (10 mL) at (0–5) °C, the hydrazone derivatives 4ad (1 mmol) were added portionwise with stirring. After addition, the cold solution was stirred for 1 h, then heated with stirring at 60–65 °C for 3h. The reaction solution was poured onto an ice–water mixture, and the acidic medium was neutralized by adding a small amount of dilute ammonia solution (10%) The formed solid was collected by filtration and recrystallized from ethanol to give the 4-formyl pyrazole derivatives 5ad.

3-(Furan-2-yl)-1-(4-oxo-3,4-dihydroquinazolin-2-yl)-1H-pyrazole-4-carbaldehyde (5a)

Yield 68%, buff powder, m.p. 191–192 °C. IR (KBr, υmax/cm−1): 3434 (NH), 3089 (CH-aromatic), 1680 (C=O), 1628 (C=N). 1H NMR (500 MHz, DMSO-d6, δ ppm): 6.67 (s, 1H, Ar-H), 7.12 (d, 1H, J = 7.6 Hz, Ar-H), 7.46–7.64 (m, 3H, Ar-H), 7.79–7.83 (m, 2H, Ar-H), 8.12 (s, 1H, CH-pyrazole), 9.68 (s, 1H, H–C=O), 10.11 (s, 1H, NH, D2O exchangeable). 13C NMR (125 MHz, DMSO-d6, δ ppm): 112.47, 113.65, 115.35, 122.77, 126.88, 127.20, 127.40, 135.52, 135.81, 136.82, 144.14, 145.31, 145.98, 153.04 (Ar-C, C=N) 162.69, 185.42 (2C=O). Anal. Calcd. for C16H10N4O3 (306.28): C, 62.74; H, 3.29; N, 18.29%; Found: C, 62.53; H, 3.14; N, 17.98%. ESI−MS: m/z = 305.22 [M-H+].

1-(4-Oxo-3,4-dihydroquinazolin-2-yl)-3-(thiophen-2-yl)-1H-pyrazole-4-carbaldehyde (5b)

Yield 67%, pale grey powder, m.p. 224 °C. IR (KBr, υmax/cm−1): 3441 (NH), 3098 (CH-aromatic), 1687 (C=O), 1617 (C=N). 1H NMR (500 MHz, DMSO-d6, δ ppm): 7.15 (d, 1H, J = 7.6 Hz, Ar-H), 7.42 (d, 1H, J = 8.6 Hz, Ar-H), 7.64–7.70 (m, 3H, Ar-H), 7.82 (d, 1H, J = 7.6 Hz, Ar-H), 8.10 (d, 1H, J = 7.7 Hz, Ar-H), 8.24 (s, 1H, CH-pyrazole) 9.44 (s, 1H, H–C=O), 10.07 (s, 1H, NH, D2O exchangeable). 13C NMR (125 MHz, DMSO-d6, δ ppm): 115.87, 122.90, 126.94, 127.19, 127.52, 127.99, 128.58, 129.55, 130.72, 133.15, 135.58, 139.12, 143.75, 153.49 (Ar-C, C=N) 162.49, 185.48 (2C=O). Anal. Calcd. for C16H10N4O2S (322.34): C, 59.62; H, 3.13; N, 17.38; S, 9.95%; Found: C, 59.87; H, 3.31; N, 17.16; S, 10.18%. ESI−MS: m/z = 321.37 [M-H+].

1-(4-Oxo-3,4-dihydroquinazolin-2-yl)-3-(pyridin-2-yl)-1H-pyrazole-4-carbaldehyde (5c)

Yield 64%, dark brown powder, m.p. 178 °C. IR (KBr, υmax/cm−1): 3404 (2NH), 3084 (CH-aromatic), 1697 (C=O), 1627 (C=N). 1H NMR (500 MHz, DMSO-d6, δ ppm): 7.12 (d, 1H, J = 7.6 Hz, Ar-H), 7.30–7.51 (m, 3H, Ar-H), 7.63–7.69 (m, 2H, Ar-H), 7.83 (d, 1H, J = 7.7 Hz, Ar-H), 8.21 (s, 1H, CH-pyrazole), 8.48 (d, 1H, J = 6.5 Hz, Ar-H) 9.53 (s, 1H, H–C=O), 10.15 (s, 1H, NH, D2O exchangeable). 13C NMR (125 MHz, DMSO-d6, δ ppm): 115.59, 122.85, 123.08, 125.21, 126.97, 127.09, 127.69, 135.10, 135.58, 136.77, 138.01, 146.54, 149.78, 153.50, 156.12 (Ar-C, C=N) 162.51, 185.39 (2C=O). Anal. Calcd. for C17H11N5O2 (317.31): C, 64.35; H, 3.49; N, 22.07%; Found: C, 64.58; H, 3.68; N, 22.29%. ESI−MS: m/z = 316.27 [M-H+].

3-(4-Methoxyphenyl)-1-(4-oxo-3,4-dihydroquinazolin-2-yl)-1H-pyrazole-4-carbaldehyde (5d)

Yield 67%, brown, m.p. 163–164 °C. IR (KBr, υmax/cm−1): 3408 (NH), 3087 (CH-aromatic), 2918 (CH-aliphatic), 1696 (C=O), 1619 (C=N). 1H NMR (500 MHz, DMSO-d6, δ ppm): 3.82 (s, 3H, OCH3), 7.13–7.25 (m, 3H, Ar-H), 7.42 (d, 1H, J = 8.6 Hz, Ar-H), 7.61–8.06 (m, 4H, Ar-H), 8.17 (s, 1H, CH-pyrazole), 9.69 (s, 1H, H–C=O), 10.06 (s, 1H, NH, D2O exchangeable). 13C NMR (125 MHz, DMSO-d6, δ ppm): 56.09 (OCH3), 112.08, 114.87, 122.91, 124.98, 126.94, 127.44, 127.65, 128.08, 129.42, 134.49, 137.33, 146.87, 153.12, 160.29 (Ar-C, C=N), 162.07, 185.31 (2C=O). Anal. Calcd. for C19H14N4O3 (346.35): C, 65.89; H, 4.07; N, 16.18%; Found: C, 65.68; H, 4.30; N, 16.47%. ESI−MS: m/z = 345.38 [M-H+].

2.2. Antimicrobial Screening

All the synthesized quinazolin-4(3H)-one compounds (2, 4af, 5ad) were screened for their in vitro antibacterial activity against four bacterial strains (Bacillus subtilis 6633, Staphylococcus aureus 25923, Salmonella typhimurium 14028, and Escherichia coli 8739) compared with Amoxicillin trihydrate as the reference drug. Furthermore, all the compounds were evaluated as antifungal agents against four fungal strains (Candida tropicals 750, Candida albicans 10231, Macrophomina phaseolina A62743, and Aspergillus niger EM77 KF774181) compared with the reference drug Clotrimazole. The diameter of inhibition zone (DIZ) assay was performed by the agar disk diffusion method [38]. The Minimum Inhibitory Concentration (MIC) of the compounds was then evaluated against bacterial strains and fungal strains using the broth dilution method [39]. (More details are provided in the Supplementary Materials S33).

2.3. DNA Gyrase Supercoiling Inhibition Assay

The assay for determining the DNA gyrase supercoiling inhibition for the target compounds (4a, 4b, 4c, 5a, 5b, 5c, and 5d) with Novobiocin as a reference inhibitor was performed according to the protocol obtained from the supplier by using the E. coli DNA gyrase kit provided by TopoGEN, Inc. (Port Orange, FL, USA) [40,41,42]. The new compounds and the standard inhibitor were dissolved in DMSO and serially diluted at concentrations of 100, 10, 1, and 0.1 μM, then assayed in reaction mixtures in three different replicate runs. The average IC50 values (μM) of the triplicate experiments were calculated for the target compounds and Novobiocin. (More details are provided in the Supplementary Materials S34).

2.4. Molecular Modeling Studies

To investigate the molecular interactions between the most potent quinazolinone derivatives 4ac and 5ad and the active site of E. coli DNA gyrase B kinase, a molecular docking study was performed by using the molecular operating environment ((MOE) software Version 2014.0901, Chemical Computing Group Inc., Montereal, Canada [43]. Then, the geometry optimization and energy minimization were applied to obtain the Conf Search module in MOE, followed by saving the MOE file for the upcoming docking process. The co-crystallized structure of E. coli DNA gyrase B kinase with its ligand novobiocin was downloaded (PDB code: 1AJ6) [44]. At first, the validation of the docking process was established by docking of the native ligand, followed by docking of the derivatives 4ac and 5ad within the ATP-binding site after elimination of co-crystallized ligand. (More details are provided in the Supplementary Materials S35; The 3D binding poses are provided in the Supplementary Materials, Figure S32).

2.5. In Silico ADME Prediction Study

The physicochemical and pharmacokinetic properties of the highly potent quinazolinone compounds 4a, 5a, 5c, and 5d were predicted by using the free web tool, SwissADME [45,46,47].

3. Results and Discussion

3.1. Chemistry

The present work included the synthesis of a series of new quinazolin-4(3H)one derivatives (4af, 5ad) as illustrated by Scheme 1 and Scheme 2. The structure of all synthesized compounds were confirmed by 1H-NMR, 13C-NMR, IR, and mass spectral data (Supplementary Materials, Figures S1–S31), in addition to the correct ratios of their elemental microanalyses. The 2-hydrazinylquinazolin-4(3H)-one (2) was reported as a key intermediate compound in the synthesis of different quinazolinone-based compounds [48,49]. For the synthesis of the target quinazolinone-hydrazone derivatives, the starting 2-hydrazinyl derivative 2 was synthesized in a good yield by the reaction of 2-thioxo-2,3-dihydroquinazolin-4(1H)-one (1) with hydrazine hydrate in refluxed ethanol. Upon treatment of the 2-hydrazinyl derivative 2 with different carbonyl compounds (3af), namely 1-(furan-2-yl)ethan-1-one, 1-(thiophen-2-yl)ethan-1-one, 1-(pyridin-2-yl)ethan-1-one, 1-(4-methoxyphenyl)ethan-1-one, 4-methylcyclohexan-1-one, and indoline-2,3-dione in glacial acetic under reflux, the hydrazone derivatives 4af was afforded (Scheme 1). The IR spectrum of the 2-hydrazinyl derivative 2 showed a strong absorption band at 1674 cm−1 related to the C=O group along with three absorption bands at the region 3431–3187 cm−1 due to the NH stretching vibration of the NH-quinazolinone and the –NHNH2 moiety. Furthermore, the 1H-NMR spectrum of 2 represented three D2O exchangeable signals in the range of δ 8.09–10.24 ppm corresponding to the protons of the two NH and the NH2 groups, besides the signals of the four quinazolinone aromatic protons at their expected regions. Moreover, the 13C-NMR data of the 2-hydrazinyl derivative 2 showed a signal at δ 163.91 ppm related to the C=O group alongside the expected signals of the C=N group and the four aromatic carbons. Moreover, the formation of the hydrazones (4af) was confirmed by their 1H-NMR spectra, which revealed vanishing of the D2O exchangeable signal of the hydrazide NH2 group at δ 8.09 ppm of the parent 2-hydrazinyl derivative 2. In addition, the 1H-NMR spectra of the arylethylidene derivatives (4ad) represented the singlet signal corresponding to the CH3 protons of the N=C–CH3 moiety in the range of δ 2.24–2.40 ppm along with the signals of the new aromatic protons related to the new aryl group. Another support to the structures of (4ad) was gained by their 13C-NMR data, which showed signals corresponding to the CH3 carbon in the range of δ 13.16–16.90 ppm besides the signals of the new aromatic carbons, while the 1H-NMR spectrum of the 4-methylcyclohexylidene derivative 4e showed the doublet signal at δ 0.88 ppm, the multiplet signal at δ 1.21–1.25 ppm, and the multiplet signal at δ 1.40–2.05 ppm assignable to the protons of CH3, CH, and 4CH2 of the new 4-methylcyclohexylidene moiety, respectively. Furthermore, the 13C-NMR of 4e assisted in showing the presence of the 4-methylcyclohexylidene carbons by five signals in the range of δ 21.75–35.55 ppm. Moreover, the IR spectrum of the 2-oxoindolin-3-ylidene derivative 4f exhibited two absorption bands at 1699 and 1671 cm−1, corresponding to the two C=O groups of the indolinone and quinazolinone moieties. Furthermore, the 1H-NMR spectrum of 4f revealed the signals corresponding to the aromatic protons of the new indolinone moiety alongside three D2O exchangeable signals assignable to quinazolinone-NH, hydrazone-NH, and the new indolinone-NH in the range of δ 10.50–11.22 ppm. Additionally, the 13C-NMR spectrum of 4f showed a new signal at δ 169.03 ppm, ascribed to indolinone-C=O, as well as new signals related to the aromatic carbons of the indolinone moiety.
Further treatment of the 2-(2-arylethylidene)hydrazinyl)quinazolin-4(3H)-ones 4af with the DMF/POCl3 mixture (Vilsmeier–Haack reagent) afforded the 3-aryl-1-(4-oxo-3,4-dihydroquinazolin-2-yl)-1H-pyrazole-4-carbaldehydes 5ad in good yields (Scheme 2), whereas the conversion of the hydrazones 4ad to the corresponding formyl-pyrazole derivatives 5ad was achieved via the reported mechanism of the Vilsmeier–Haack reaction [50]. The IR spectra of the formyl-pyrazole derivatives 5ad showed one strong absorption band in the region 1697–1680 cm−1, corresponding to the two C=O groups of the quinazolinone and the formyl-pyrazole moieties. The 1H-NMR spectra of 5ad showed only one D2O exchangeable signal in the range of δ 10.07–10.15 ppm corresponding to quinazolinone-NH with vanishing of the ethylidene-CH3 signal, which confirmed the formation of the formyl-pyrazole moiety. Moreover, the 1H-NMR spectra of 5ad revealed, besides the signals of the parent aromatic protons, two new singlet signals at ranges δ 8.12–8.24 ppm and δ 9.44–9.69 ppm, ascribed to CH-pyrazole and the H–C=O protons, respectively. Furthermore, the 13C-NMR spectra 5ad showed the signals related to all the carbons in the expected regions alongside the signal of the H–C=O carbon in the range of δ 185.31–185.48 ppm. In addition, the molecular structures of the new compounds (4af, 5ad) were confirmed by their mass spectra, which represent their correct molecular ion peaks.

3.2. Antimicrobial Activity

All the synthesized quinazolin-4(3H)one derivatives (2, 4af, 5ad) were examined as antimicrobial agents against a number of bacterial and fungal strains. The tested microorganisms include two Gram-positive bacteria (B. subtilis 6633, S. aureus 25923), two Gram-negative bacteria (S. typhimurium 14028, E coli 8739), two yeast strains (C. tropicals 750, C. albicans 10231), and two fungal strains (M. phaseolina A62743, A. niger EM77-KF774181) compared with Amoxicillin trihydrate and Clotrimazole as reference drugs against the bacterial and fungal strains, respectively. The average diameters of the inhibition zones in mm were determined for the target compounds and the reference antibiotics and are listed in Table 1. Then, the MIC values in (μg/mL) were determined and are listed in Table 2 and represented by Figure 2 and Figure 3.
The obtained results of the inhibition zones of the new quinazolin-4(3H)-one derivatives (Table 1) revealed that some of the new compounds such as 4a, 4c, 5a, 5c, and 5d exhibited large inhibition zones ranging from 22 to 39 mm against bacterial and fungal strains, which indicates the higher sensitivity of the tested strains to the target compounds.
According to the MIC values, it is obvious that the majority of the target compounds showed significant activity against the tested microbial strains. 2-hydrazinylquinazolin-4(3H)-one (2) revealed antimicrobial activity varying from moderate to weak with MIC values in range of (16–128) μg/mL, while the target hydrazone derivatives (4af) showed a wide variety in their potency against both bacterial and fungal strains, whereas the most potent antibacterial activity was exhibited by the 2-(1-(furan-2-yl)ethylidene derivative 4a, which exceeded the potency of Amoxicillin against E. coli with a MIC value 4 μg/mL (MICAmoxxicillin = 8 μg/mL) and showed equipotent activity to that of Amoxicillin against S. aureus, B. subtilis, and S. typhimurium with MIC values of 4, 4, and 8 μg/mL, respectively. Furthermore, 4a showed potent antifungal activity with MIC values in the range of (2–16) μg/mL, which is more potent than Clotrimazole against C. albicans and M. phaseolina with MIC values of 2 and 8 μg/mL compared with the MIC values of 8 and 16 μg/mL of Clotrimazole, respectively. Moreover, the 2-(1-(pyridin-2-yl)ethylidene derivative 4c showed more potent antibacterial activity than Amoxicillin against S. typhimurium with a MIC value of 4 μg/mL and showed equipotent activity to that of Amoxicillin against E. coli and S. typhimurium with a MIC value of 8 μg/mL. Additionally, the antifungal activity of 4c was the most potent between the target hydrazones with MIC values in the range of (2–8) μg/mL, which exceeded the antifungal activity of Clotrimazole against the four tested strains. The activity of the 2-(1-(thiophen-2-yl)ethylidene derivative 4b came after 4a and 4c against bacterial strains; it showed similar sensitivity towards the four tested strains with a MIC value of 8 μg/mL. However, it revealed lower antifungal activity with MIC values in the range of (16–128) μg/mL. Furthermore, the 2-(4-methylcyclohexylidene derivative 4e showed moderate activity against the tested microorganisms with MIC values varying from 16 to 32 μg/mL. An apparent decrease of the antimicrobial activity was shown by the 2-(1-(4-methoxyphenyl)ethylidene derivative 4d and 2-oxoindolin-3-ylidene derivative 4f; they revealed weak or no activity against the tested bacterial and fungal strains.
In addition, the conversion of the hydrazone derivatives 4ad to the formyl-pyrazole derivatives 5ad led to an obvious increase in the antimicrobial activity, especially against the bacterial strains, whereas the 3-(furan-2-yl)-1H-pyrazole-4-carbaldehyde derivative 5a exhibited the most potent antimicrobial activity among the tested compounds and the reference drugs with MIC values in the range of (1–4) μg/mL. Moreover, the 3-(thiophen-2-yl)-1H-pyrazole-4-carbaldehyde 5b showed similar antibacterial activity to that of Amoxicillin against three of the tested bacterial strains and showed moderate antifungal activity against the four fungal strains with the same MIC value = 32 μg/mL. In addition, the 3-(pyridin-2-yl)-1H-pyrazole-4-carbaldehyde derivative 5c displayed more potent activity than that of Amoxicilli against E. coli and S. typhimurium with MIC values of 2 and 4 μg/mL, respectively, while it showed some decrease in the activity against the four tested fungal strains, compared with its parent hydrazone derivative 4c, with MIC values in the range of (8–16) μg/mL. Finally, the 3-(4-methoxyphenyl)-1H-pyrazole-4-carbaldehyde 5d showed much higher potency against both the tested bacterial and fungal strains compared with 4d. It revealed more potent activity than Amoxicillin against E. coli and equal potency to the reference drugs against the other tested microorganisms.

3.3. In Vitro DNA Gyrase Inhibitory Activity

The target compounds of the most potent antibacterial activity (4a, 4b, 4c, 5a, 5b, 5c, and 5d) were subjected to further screening for their inhibition profiles against E coli DNA gyrase compared with Novobiocin as a reference DNA gyrase inhibitor. The results of the IC50 values of the tested compounds in Table 3 showed that the most potent inhibitory activity was exhibited by the 3-(furan-2-yl)-1H-pyrazole-4-carbaldehyde derivative 5a with an IC50 value of 3.19 µM and the 3-(4-methoxyphenyl)-1H-pyrazole-4-carbaldehyde 5d with an IC50 value of 3.51 µM, which exceeded the inhibition potency of Novobiocin with an IC50 value of 4.12 µM. Furthermore, the 3-(pyridin-2-yl)-1H-pyrazole-4-carbaldehyde derivative 5c and the 2-(1-(furan-2-yl)ethylidene derivative 4a showed potent inhibition with IC50 values of 4.09 and 4,17 µM, respectively, which were nearly equal to that of the reference inhibitor, while the other target compounds 4b, 4c, and 5b revealed lower inhibition potency against the target enzyme with IC50 values in the range of (10.82–16.91) µM.

3.4. Molecular Docking Study on E. coli DNA Gyrase B Kinase

The docking simulation of the most active novel quinazolin-4(3H)one derivatives (4ac, 5ad) was established in an attempt to explain their variable gained potencies with E. coli DNA gyrase B kinase. The process of molecular docking was firstly validated through self-docking of the co-crystallized ligand Novobiocin within the active site of E. coli DNA gyrase B (PDB code: 1AJ6), giving an energy score of −10.25 kcal/mol with small RMSD values of 0.87 Å between the native ligand and its docked pose. The docking results are inserted in Table 4 and revealed that the derivatives 4ac and 5ad occupied the same binding site through two types of interactions: one was arene–cation interactions between the quinazolinone moiety and Ile78, and the other was between the oxygen of the quinazolinone and Gly77, whereas the highest binding affinity was revealed by the target compounds 4a, 5a, 5c, and 5d, which exhibited the most potent inhibition of E. coli DNA gyrase, with the energy score ranging from −9.89 to −10.97 kcal/mol, while the other compounds 4b, 4c, and 5b showed lower binding affinity with the energy score ranging from −8.66 to −8.80 kcal/mol.
By inspection of Figure 4, which describes the binding manner of the highly potent compounds 4a, 5a, 5c, and 5d, it was observed that the hydrazineyl derivative 4a exhibited three additional H-bonds with the sidechain of Asp73 (distance: 2.74, 3.08, and 3.16 Å, respectively). Moreover, the furanyl moiety displayed arene–cation interaction with key amino acid Asn46, which was not shown with the corresponding thienyl and pyridinyl moieties in the derivatives 4b and 4c. Furthermore, these moieties forced the hydrazineyl group away from binding with the essential amino acid Asp73. The formation of the formylpyrazole scaffold in the derivatives 5a, 5c, and 5d gave the chance for good fitting through H-bonding or arene–cation interaction with Asn46.
According to the superimposition between the promising new compounds 4a, 5a, 5c, and 5d (Figure 5), the existence of the quinazolinone scaffold facilitated binding within the active site of E. coli DNA gyrase B through hydrophilic and hydrophobic interactions. Furthermore, incorporation with furanyl-hydrazone and formylpyrazole cores increased the chance for more fitting, and that could be a lead for the discovery of new E. coli DNA gyrase B inhibitors.

3.5. In Silico ADME Study

The study of absorption, distribution, metabolism, and excretion (ADME) for the designed targets is a key insight to obtain the optimal drug candidate. This prediction study was afforded using the free online tool, SwissADME. The optimum orally available drug obeys Lipinski’s rule (molecule with MW ≤ 500, number of hydrogen bond donors ≤ 5, number of hydrogen bond acceptors ≤ 10, and MLogP ≤ 4.15) and Veber’s rule (TPSA ≤ 140 Å2, number of rotatable bonds ≤ 10). It was observed that all the assessed targets 4a, 5a, 5c, and 5d were compatible with the previous rules with no violations (Table 5). Concerning the bioavailability radar chart (Figure 6), it was noted that all compounds were located in the preferred range (pink area) corresponding to the five parameters lipophilicity (LIPO), flexibility (FLEX), solubility (INSOLU), polarity (POLAR), and SIZE, but kept away from the desirable area of saturation (INSATU), and that represented good prediction for their oral bioavailability.
The pharmacokinetic properties of the promising new quinazolin-4(3H)one derivatives 4a, 5a, 5c, and 5d were investigated and are depicted in Table 6 and Figure 7. All the derivatives revealed high expected gastrointestinal absorption with no BBB penetration, as they all inserted in the white region of the boiled-egg chart and away from the yellow one. Therefore, they were not predicted to cause CNS side effects and can be applied only for peripheral infections. It is known that p-glycoprotein (P-gp) is a drug efflux transporter and responsible for the pumping of drugs out of the cell, which could be one of the reasons for drug resistance. Through the SwissADME website, it was expected that all the screened compounds were not substrates for P-gp (red points, Figure 7), signifying a low chance of their efflux out of the cell with a maximum activity. Moreover, these derivatives exhibited high bioavailability scores of 0.55 with no PAIN alert.

4. Conclusions

The present study involved the design and synthesis of quinazolin-4(3H)-one derivatives (4af, 5ad) bearing hydrazone or formyl-pyrazole moieties as new antimicrobial agents targeting DNA gyraze enzyme. All the target quinazolin-4(3H)-one derivatives were evaluated for their in vitro antibacterial activities compared with Amoxicillin trihydrate against two Gram-positive bacteria and two Gram-negative bacteria. Furthermore, the target compounds were evaluated for their in vitro antifungal activities compared with Clotrimazole as the reference drug against two yeasts and two fungi. The new quinazolin-4(3H)-one derivatives showed significant antimicrobial activity compared with the reference drugs, especially the formyl-pyrazole derivatives. The 3-(Furan-2-yl)-1H-pyrazole-4-carbaldehyde derivative 5a showed the most potent activity between the target compounds and the reference drugs with MIC values in the range of (1–16) μg/mL. In addition, the most active compounds (4a, 4b, 4c, 5a, 5b, 5c, and 5d) were evaluated for their inhibition activity against E. coli DNA gyrase compared with Novobiocin as the reference inhibitor. The results revealed that the most potent inhibition was achieved by 5a with an IC50 value of 3.19 µM compared with Novobiocin with an IC50 value of 4.12 µM. Furthermore, the formyl pyrazole derivatives 5c and 5d showed potent inhibition activity of the target compounds with IC50 values of 4.09 and 3.51 µM, respectively. Moreover, the 2-(1-(furan-2-yl)ethylidene derivative 4a was the most potent hydrazone derivative against DNA gyrase with an IC50 value of 4,17 µM, which sheds light on the role of the furanyl moiety in enhancing the antimicrobial activity and the DNA gyrase inhibitory activity. In addition, molecular docking studies were performed for the most active target quinazolin-4(3H)-one derivatives to predict their binding mode in the target E. coli DNA gyrase B active site. The results of the docking study were compatible with the DNA gyrase inhibitory activities, where the most binding affinity was obtained by the most potent inhibitors (4a, 5a, 5c, and 5d) with the energy score range of (−9.89–−10.97) kcal/mol. Furthermore, ADME studies of the most active compounds predicted their good oral bioavailability.
The results gained from the present work revealed the significant antimicrobial activity of the target quinazolin-4(3H)-one compounds, especially compounds 4a, 5a, 5c, and 5d, as broad-spectrum antimicrobial agents targeting DNA gyrase enzyme.

Supplementary Materials

The following Supporting Information can be downloaded at: https://www.mdpi.com/article/10.3390/scipharm90030052/s1, Figures S1–S31: all the NMR spectra and some of the I.R and MS spectra of the new compounds; Figure S32: the 3D binding poses of compounds 4a, 5a, 5c, and 5d within the active site of E. coli DNA gyrase B; S33: in vitro antimicrobial assay; S34: in vitro DNA gyrase inhibitory assay; S35: molecular docking studies.

Author Contributions

Conceptualization, E.M.M.E.-D.; investigation, E.M.M.E.-D. and E.A.K.; methodology, E.M.M.E.-D. and E.A.K.; software, E.S.N.; writing—original draft preparation, E.M.M.E.-D. and E.S.N.; writing—review and editing, E.M.M.E.-D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data are mentioned in the manuscript.

Acknowledgments

The authors thank the National Research Centre for supporting this work.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sifri, Z.C.; Chokshi, A.; Cennimo, D.; Horng, H. Global Contributors to Antibiotic Resistance. J. Glob. Infect. Dis. 2019, 11, 36–42. [Google Scholar] [CrossRef] [PubMed]
  2. Lomazzi, M.; Moore, M.; Johnson, A.; Balasegaram, M.; Borisch, B. Antimicrobial resistance—Moving forward? BMC Public Health 2019, 19, 858. [Google Scholar] [CrossRef] [PubMed]
  3. Li, B.; Webste, T.J. Bacteria antibiotic resistance: New challenges and opportunities for implant-associated orthopedic infections. J. Orthop. Res. 2018, 36, 22–32. [Google Scholar] [CrossRef] [PubMed]
  4. Teitzel, G. Responding to Antimicrobial Resistance with Novel Therapeutics. Trends Microbiol. 2019, 27, 285–286. [Google Scholar] [CrossRef] [PubMed]
  5. Egorov, A.M.; Ulyashova, M.M.; Rubtsova, M.Y. Bacterial Enzymes and Antibiotic Resistance. Acta Naturae 2018, 10, 33–48. [Google Scholar] [CrossRef] [PubMed]
  6. Dubey, K.K.; Sharma, M. Reprogramming of antibiotics to combat antimicrobial resistance. Arch. Pharm. 2020, 353, e2000168. [Google Scholar] [CrossRef]
  7. Klostermeier, D. Why Two? On the Role of (A-)Symmetry in Negative Supercoiling of DNA by Gyrase. Int. J. Mol. Sci. 2018, 19, 1489. [Google Scholar] [CrossRef]
  8. Ma, Z.; Mduli, K. Mycobacterium tuberculosis DNA Gyrase as a Target for Drug Discovery. Infect. Disord. Drug Targets 2007, 7, 159–168. [Google Scholar] [CrossRef]
  9. Annunziato, G. Strategies to Overcome Antimicrobial Resistance (AMR) Making Use of Non-Essential Target Inhibitors: A Review. Int. J. Mol. Sci. 2019, 20, 5844. [Google Scholar] [CrossRef]
  10. Yi, L.; Lu, X. New Strategy on Antimicrobial-resistance: Inhibitors of DNA Replication Enzymes. Curr. Med. Chem. 2019, 26, 1761–1787. [Google Scholar] [CrossRef] [PubMed]
  11. Yang, H.; Li, Q.; Su, M.; Luo, F.; Liu, Y.; Wang, D.; Fan, Y. Design, synthesis, and biological evaluation of novel 6-(pyridin-3-yl) quinazolin-4(3H)-one derivatives as potential anticancer agents via PI3K inhibition. Bioorg. Med. Chem. 2021, 46, 116346. [Google Scholar] [CrossRef] [PubMed]
  12. Mirgany, T.O.; Abdalla, A.N.; Arifuzzaman, M.; Motiur Rahman, A.F.M.; Al-Salem, H.S. Quinazolin-4(3H)-one based potential multiple tyrosine kinase inhibitors with excellent cytotoxicity. J. Enzyme Inhib Med. Chem. 2021, 36, 2055–2067. [Google Scholar] [CrossRef] [PubMed]
  13. Ramadan, S.K.; Elrazaz, E.Z.; Abouzid, K.A.M.; El-Naggar, A.M. Design, synthesis and in silico studies of new quinazolinone derivatives as antitumor PARP-1 inhibitors. RSC Adv. 2020, 10, 29475–29492. [Google Scholar] [CrossRef] [PubMed]
  14. Masri, A.; Anwar, A.; Khan, N.A.; Shahbaz, M.S.; Khan, K.M.; Shahabuddin, S.; Siddiqui, R. Antibacterial Effects of Quinazolin-4(3H)-One Functionalized-Conjugated Silver Nanoparticles. Antibiotics 2019, 8, 179. [Google Scholar] [CrossRef] [PubMed]
  15. Kavitha, K.; Mohan, S.; Srinivasan, N.; Suresh, R. Design, Synthesis, Characterization of Antimicrobial activity of Schiff bases of novel Quinazolin-4-one Derivatives. Res. J. Pharm. Tech. 2021, 14, 466–470. [Google Scholar] [CrossRef]
  16. Demirel, U.U.; Yilmaz, A.; Türkdağı, H.; Öztürk, B.; Arslan, U. Investigation of Some 3H-Quinazolin-4-One Derivatives in Vitro Antimicrobial Effect and Cytotoxicity on Human Gingival Fibroblasts. Polycycl. Aromat. Compd. 2021, 41, 1623–1630. [Google Scholar] [CrossRef]
  17. Thorat, D.B.; Shivkumar, B.; Rao, N.; Mohankumar, K.M. Synthesis and Evaluation of new 4(3H)-quinazolinones derivatives as potential Anti-Inflammatory agents. Asian J. Pharm. Sci. 2021, 11, 213–218. [Google Scholar] [CrossRef]
  18. Kumar, S.; Aghara, J.C.; Alex, A.T.; Aranjani, J.M.; Joesph, A. Novel Quinolone Substituted Quinazolin-4(3H)-Ones as Anti-Inflammatory, Anticancer Agents: Synthesis and Biological Screening. Indian J. Pharm. Educ. Res. 2018, 52, S268–S276. [Google Scholar] [CrossRef]
  19. Ayyad, R.A.; Sakr, H.M.; El-Gamal, K.M. Design, Synthesis, Computer Modeling and Analgesic Activity of Some New Disubstituted Quinazolin-4(3H)-ones. Med. Chem. 2016, 6, 299–305. [Google Scholar] [CrossRef]
  20. Osarumwense, P.; Edema, M.; Usifoh, C. Synthesis and Anagesic activities of Quinazolin-4(3H)-One, 2-Methyl-4(3H)-Quinazolinone and 2–Phenyl-4(3H)-quinazolin-4(3H)–one. J. Drug Deliv. Ther. 2020, 10, 87–91. [Google Scholar] [CrossRef]
  21. Birhan, Y.S.; Bekhit, A.A.; Hymete, A. In vivo antimalarial evaluation of some 2,3-disubstituted-4(3H)-quinazolinone derivatives. BMC Res. Notes 2015, 8, 589. [Google Scholar] [CrossRef]
  22. Bule, M.H.; Haymete, A.; Kefale, B. Synthesis and In-Vivo Pharmacological Evaluation of Some Novel 4(3H)- Quinazolinone Derivatives as Potential Anti-malarial Agents. Drug. Des. 2015, 4, 121. [Google Scholar] [CrossRef]
  23. Chen, L.; Wang, X.; Tang, X.; Xia, R.; Guo, T.; Zhang, C.; Li, X.; Xue, W. Design, synthesis, antiviral bioactivities and interaction mechanisms of penta-1,4-diene-3-one oxime ether derivatives containing a quinazolin-4(3H)-one scaffold. BMC Chem. 2019, 13, 34. [Google Scholar] [CrossRef]
  24. Tran, T.P.; Ellsworth, E.L.; Sanchez, J.P.; Watson, B.M.; Stier, M.A.; Showalter, H.D.H.; Domagala, J.M.; Shapiro, M.A.; Joannides, E.T.; Gracheck, S.J.; et al. Structure–activity relationships of 3-aminoquinazolinediones, a new class of bacterial type-2 topoisomerase (DNA gyrase and topo IV) inhibitors. Bioorg. Med. Chem. Lett. 2007, 17, 1312. [Google Scholar] [CrossRef]
  25. Ghorab, M.M.; Alqahtani, A.S.; Soliman, A.M.; Askar, A.A. Novel N-(Substituted) Thioacetamide Quinazolinone Benzenesulfonamides as Antimicrobial Agents. Int. J. Nanomed. 2020, 15, 3161–3180. [Google Scholar] [CrossRef]
  26. Xue, W.; Wang, Y.; Lian, X.; Li, X.; Pang, J.; Kirchmair, J.; Wu, K.; Han, Z.; You, X.; Zhang, H.; et al. Discovery of N-quinazolinone-4-hydroxy-2-quinolone-3-carboxamides as DNA gyrase B-targeted antibacterial agents. J. Enzyme Inhib. Med. Chem. 2022, 37, 1620–1631. [Google Scholar] [CrossRef]
  27. Verma, G.; Marella, A.; Shaquiquzzaman, M.; Akhtar, M.; Ali, M.R.; Alam, M.M. A review exploring biological activities of hydrazones. J. Pharm. Bioallied Sci. 2014, 6, 69–80. [Google Scholar] [CrossRef]
  28. Rollas, S.; Küçükgüzel, S.G. Biological Activities of Hydrazone Derivatives. Molecules 2007, 12, 1910–1939. [Google Scholar] [CrossRef]
  29. Ansari, A.; Ali, A.; Asif, A. Shamsuzzman Review: Biologically active pyrazole derivatives. New J. Chem. 2017, 41, 16–41. [Google Scholar] [CrossRef]
  30. Karrouchi, K.; Radi, S.; Ramli, Y.; Taoufik, J.; Mabkhot, Y.N.; Al-aizari, F.A.; Ansar, M. Synthesis and Pharmacological Activities of Pyrazole Derivatives: A Review. Molecules 2018, 23, 134. [Google Scholar] [CrossRef] [Green Version]
  31. Sridhar, P.; Alagumuthu, M.; Arumugam, S.; Reddy, S.R. Synthesis of quinoline acetohydrazide-hydrazone derivatives evaluated as DNA gyrase inhibitors and potent antimicrobial agents. RSC Adv. 2016, 6, 64460–64468. [Google Scholar] [CrossRef]
  32. Ragab, A.; Elsisi, D.M.; Abu Ali, O.A.; Abusaif, M.S.; Askar, A.A.; Farag, A.A.; Ammar, Y.A. Design, synthesis of new novel quinoxalin-2(1H)-one derivatives incorporating hydrazone, hydrazine, and pyrazole moieties as antimicrobial potential with in-silico ADME and molecular docking simulation. Arab. J. Chem. 2022, 15, 103497. [Google Scholar] [CrossRef]
  33. Hassan, A.S.; Askar, A.A.; Naglah, A.M.; Almehizia, A.A.; Ragab, A. Discovery of New Schiff Bases Tethered Pyrazole Moiety: Design, Synthesis, Biological Evaluation, and Molecular Docking Study as Dual Targeting DHFR/DNA Gyrase Inhibitors with Immunomodulatory Activity. Molecules 2020, 25, 2593. [Google Scholar] [CrossRef]
  34. Sun, J.; Lv, P.C.; Yin, Y.; Yuan, R.J.; Ma, J.; Zhu, H.L. Synthesis, structure and antibacterial activity of potent DNA gyrase inhibitors: N’-benzoyl-3-(4-bromophenyl)-1H-pyrazole-5-carbohydrazide derivatives. PLoS ONE 2013, 8, e69751. [Google Scholar] [CrossRef]
  35. Viegas-Junior, C.; Danuello, A.; da Silva Bolzani, V.; Barreiro, E.J.; Fraga, C.A. Molecular hybridization: A useful tool in the design of new drug prototypes. Curr. Med. Chem. 2007, 14, 1829–1852. [Google Scholar] [CrossRef] [PubMed]
  36. Sree, B.S.; Rao, A.L.; Babu, P.S.; Kiran, K.S.; Vandana, K. Synthesis and antimicrobial screening of some novel quinazolinones and its derivatives. Indian J. Pharm. Pharmacol. 2017, 4, 130–133. [Google Scholar] [CrossRef]
  37. Gruner, M.; Rehwald, M.; Katrin Eckert, K.; Gewald, K. New Syntheses of 2-Alkylthio-4-Oxo-3,4-Dihydroquinazolines, 2-Alkylthioquinazolines, as Well as Their Hetero Analogues. Heterocycles 2000, 53, 2363–2377. [Google Scholar] [CrossRef]
  38. Penna, C.A.; Marino, S.G.; Gutkind, G.O.; Clavin, M.; Ferraro, G.; Martino, V. Antimicrobial activity of Eupatorium species growing in Argentina. J. Herbs Spices Med. Plants 1988, 5, 21–28. [Google Scholar] [CrossRef]
  39. Wiegand, I.; Hilpert, K.; Hancock, R.E. Agar and broth dilution methods to determine the minimal inhibitory concentration (MIC) of antimicrobial substances. Nat. Protoc. 2008, 3, 163–175. [Google Scholar] [CrossRef]
  40. Phillips, J.W.; Goetz, M.A.; Smith, S.K.; Zink, D.L.; Polishook, J.; Onishi, R.; Salowe, S.; Wiltsie, J.; Allocco, J.; Sigmund, J.; et al. Discovery of Kibdelomycin, A Potent New Class of Bacterial Type II Topoisomerase Inhibitor by Chemical-Genetic Profiling in Staphylococcus aureus. Chem. Biol. 2011, 18, 955–965. [Google Scholar] [CrossRef] [Green Version]
  41. Maxwell, A.; Burton, N.P.; O’Hagan, N. High-throughput assays for DNA gyrase and other topoisomerases. Nucleic Acids Res. 2006, 34, e104. [Google Scholar] [CrossRef]
  42. Mohi El-Deen, E.M.; Abd El-Meguid, E.A.; Karam, E.A.; Nossier, E.S.; Ahmed, M.F. Synthesis and Biological Evaluation of New Pyridothienopyrimidine Derivatives as Antibacterial Agents and Escherichia coli Topoisomerase II Inhibitors. Antibiotics 2020, 9, 695. [Google Scholar] [CrossRef]
  43. Hashem, H.E.; Amr, A.E.G.E.; Nossier, E.S.; Elsayed, E.A.; Azmy, E.M. Synthesis, antimicrobial activity and molecular docking of novel thiourea derivatives tagged with thiadiazole, imidazole and triazine moieties as potential DNA gyrase and topoisomerase IV inhibitors. Molecules 2020, 25, 2766. [Google Scholar] [CrossRef]
  44. Mohi El-Deen, E.M.; El-Meguid, E.A.A.; Hasabelnaby, S.; Karam, E.A.; Nossier, E.S.; El-Deen, E.M.; El-Meguid, E.A.A. Synthesis, Docking Studies, and In Vitro Evaluation of Some Novel Thienopyridines and Fused Thienopyridine-Quinolines as Antibacterial Agents and DNA Gyrase Inhibitors. Molecules 2019, 24, 3650. [Google Scholar] [CrossRef]
  45. 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]
  46. Othman, I.M.; Alamshany, Z.M.; Tashkandi, N.Y.; Gad-Elkareem, M.A.; Anwar, M.M.; Nossier, E.S. New pyrimidine and pyrazole-based compounds as potential EGFR inhibitors: Synthesis, anticancer, antimicrobial evaluation and computational studies. Bioorg. Chem. 2021, 114, 105078. [Google Scholar] [CrossRef]
  47. Mohi El-Deen, E.M.; Anwar, M.M.; Abd El-Gwaad, A.A.; Karam, E.A.; El-Ashrey, M.K.; Kassab, R.R. Design and synthesis of some novel pyridothienopyrimidine derivatives and their biological evaluation as antimicrobial and anticancer agents targeting EGFR enzyme. Arab. J. Chem. 2022, 15, 103751. [Google Scholar] [CrossRef]
  48. El-Tombary, A.A.; Ismail, K.A.; Aboulwafa, O.M.; Omar, A.M.M.; El-Azzouni, M.Z.; El-Mansoury, S.T. Novel triazolo [4, 3-a] quinazolinone and bis-triazolo [4, 3-a: 4, 3′-c] quinazolines: Synthesis and antitoxoplasmosis effect. Farmaco 1999, 54, 486–495. [Google Scholar] [CrossRef]
  49. Rafeeq, M.; Reddy, B.S.; Reddy, C.V.R.; Naidu, A.; Dubey, P.K. Green and efficient synthesis of 2-(4-oxo-3, 4-dihydroquinazolin-2-yl)-2, 3-dihydropthalazine-1, 4-dione. Indian J. Chem. 2015, 54B, 412–417. [Google Scholar] [CrossRef]
  50. Singh, K.; Ralhan, S.; Sharma, P.K.; Dhawan, S.N. Vilsmeier–Haack Reaction on Hydrazones: A Convenient Synthesis of 4-formylpyrazoles. J. Chem. Res. 2005, 2005, 316–318. [Google Scholar] [CrossRef]
Figure 1. Reported quinazolin-4(3H)one derivatives (I-III), hydrazone derivatives (IV,V), and pyrazole compounds (VI,VII) as antimicrobial agents targeting DNA gyrase and the design of the target compounds.
Figure 1. Reported quinazolin-4(3H)one derivatives (I-III), hydrazone derivatives (IV,V), and pyrazole compounds (VI,VII) as antimicrobial agents targeting DNA gyrase and the design of the target compounds.
Scipharm 90 00052 g001
Scheme 1. Synthesis of new quinazolin-4(3H)one derivatives bearing different hydrazone moieties (4af).
Scheme 1. Synthesis of new quinazolin-4(3H)one derivatives bearing different hydrazone moieties (4af).
Scipharm 90 00052 sch001
Scheme 2. Synthesis of new quinazolin-4(3H)one derivatives bearing 4-formyl-pyrazoles (5ad).
Scheme 2. Synthesis of new quinazolin-4(3H)one derivatives bearing 4-formyl-pyrazoles (5ad).
Scipharm 90 00052 sch002
Figure 2. The antibacterial activity (MIC in µg/mL) of the of the synthesized quinazolin-4(3H)one derivatives compared with Amoxicillin trihyderate.
Figure 2. The antibacterial activity (MIC in µg/mL) of the of the synthesized quinazolin-4(3H)one derivatives compared with Amoxicillin trihyderate.
Scipharm 90 00052 g002
Figure 3. The antifungal activity (MIC in µg/mL) of the synthesized quinazolin-4(3H)one derivatives compared with Clotrimazole.
Figure 3. The antifungal activity (MIC in µg/mL) of the synthesized quinazolin-4(3H)one derivatives compared with Clotrimazole.
Scipharm 90 00052 g003
Figure 4. (AD) Maps illustrate the 2D-binding poses of the promising quinazolinone targets, 4a, 5a, 5c, and 5d within the active site of E. coli DNA gyrase B (PDB code: 1AJ6).
Figure 4. (AD) Maps illustrate the 2D-binding poses of the promising quinazolinone targets, 4a, 5a, 5c, and 5d within the active site of E. coli DNA gyrase B (PDB code: 1AJ6).
Scipharm 90 00052 g004
Figure 5. The 3D superimposition between the most potent compounds, 4a (green), 5a (blue), 5c (yellow), and 5d (purple), and the docked original ligand, Novobiocin (red), within the binding site of E. coli DNA gyrase B (PDB code: 1AJ6).
Figure 5. The 3D superimposition between the most potent compounds, 4a (green), 5a (blue), 5c (yellow), and 5d (purple), and the docked original ligand, Novobiocin (red), within the binding site of E. coli DNA gyrase B (PDB code: 1AJ6).
Scipharm 90 00052 g005
Figure 6. The bioavailability radar chart illustrating the oral bioavailability parameters of compounds 4a, 5a, 5c, and 5d.
Figure 6. The bioavailability radar chart illustrating the oral bioavailability parameters of compounds 4a, 5a, 5c, and 5d.
Scipharm 90 00052 g006
Figure 7. Boiled-egg chart illustrating the pharmacokinetic properties of the promising new quinazolin-4(3H)one derivatives.
Figure 7. Boiled-egg chart illustrating the pharmacokinetic properties of the promising new quinazolin-4(3H)one derivatives.
Scipharm 90 00052 g007
Table 1. In vitro antimicrobial activities of the synthesized quinazolin-4(3H)-one derivatives represented as the mean diameter of the inhibition zone in mm.
Table 1. In vitro antimicrobial activities of the synthesized quinazolin-4(3H)-one derivatives represented as the mean diameter of the inhibition zone in mm.
Antibacterial ActivityAntifungal Activity
Compd.S.
aureus
B.
subtilis
E.
coli
S.
typhimurium
C.
albicans
C.
tropicals
M.
phaseolina
A.
niger
22522201420201919
4a3029302732252725
4b2624262620181626
4c2928282930322930
4d1718201615141518
4e2422212220222220
4f151716--14-17
5a3534353839353029
5b2624262616212019
5c3030333127282422
5d3129342628292527
Amoxicillin29282827----
Clotrimazole----26252426
Table 2. In vitro antimicrobial activities of the synthesized quinazolin-4(3H)-one derivatives as minimum inhibitory concentrations (MIC) in μg/mL.
Table 2. In vitro antimicrobial activities of the synthesized quinazolin-4(3H)-one derivatives as minimum inhibitory concentrations (MIC) in μg/mL.
Antibacterial ActivityAntifungal Activity
Compd.S.
aureus
B.
subtilis
E.
coli
S.
typhimurium
C.
albicans
C.
tropicals
M.
phaseolina
A.
niger
216326412832326432
4a444828816
4b8888326412816
4c48842484
4d64646464128128128128
4e1632321632161632
4f12864128--128-64
5a22111244
5b488832323232
5c4424881616
5d442888168
Amoxicillin4488----
Clotrimazole----88168
Table 3. In vitro enzymatic inhibitory activity against E coli DNA gyrase.
Table 3. In vitro enzymatic inhibitory activity against E coli DNA gyrase.
Compound No.DNA Gyrase Supercoiling Inhibition
IC50 (µM)
4a4.17 ± 0.07
4b16.91 ± 0.36
4c15.08 ± 0.34
5a3.19 ± 0.06
5b10.82 ± 0.24
5c4.09 ± 0.13
5d3.51 ± 0.10
Novobiocin4.12 ± 0.11
Table 4. Molecular docking results of the most active quinazolin-4(3H)one derivatives with E. coli DNA gyrase.
Table 4. Molecular docking results of the most active quinazolin-4(3H)one derivatives with E. coli DNA gyrase.
Compd.Docking Score
(Kcal/mol)
Amino Acid Residues
(Bond Length A°)
Atoms of CompoundType of Bond
Novobiocin−10.25Asn46(3.27);H(OH)(oxan-4-yl);H-don
Asp73(1.91);H(OCONH2);H-don
Arg76C6H2(coumarin)Arene–cation
4a−10.48Asn46;furan;Arene–cation
Asp73(3.08,3.16);NH(quinazolinone)H-don
Asp73(2.74);NH(hydrazinyl)H-don
Gly77(2.63);O(quinazolinone)H-acc
Ile78QuinazolinoneArene–cation
4b−8.70Gly77(2.66);O(quinazolinone)H-acc
Ile78QuinazolinoneArene–cation
4c−8.66Gly77(2.83);O(quinazolinone)H-acc
Ile78QuinazolinoneArene–cation
5a−10.97Asn46(2.53);O(CHO)H-acc
Gly77(2.68);O(quinazolinone)H-acc
Ile78QuinazolinoneArene–cation
5b−8.80Gly77(2.51);O(quinazolinone)H-acc
Ile78QuinazolinoneArene–cation
5c−9.89Asn46;PyrazoleArene–cation
Ile78;PyrazoleArene–cation
Ile78QuinazolinoneArene–cation
5d−10.74Asn46(2.44);O(CHO)H-acc
Asn46;4-methoxyphenylArene–cation
Ile78;PyrazoleArene–cation
Ile78QuinazolinoneArene–cation
Table 5. Predicted physicochemical properties of the most active quinazolin-4(3H)one derivatives 4a, 5a, 5c, and 5d.
Table 5. Predicted physicochemical properties of the most active quinazolin-4(3H)one derivatives 4a, 5a, 5c, and 5d.
Compd.MW 1nHBD 2nHBA 3nRB 4MLogP 5TPSA (Å2) 6Violations 7
4a268.272431.5883.280
5a306.281531.3893.780
5c317.301531.5893.530
5d346.341542.0589.870
1 Molecular weight; 2 number of hydrogen bond donor; 3 number of hydrogen bond acceptors; 4 number of rotatable bonds; 5 calculated lipophilicity (MLog Po/w); 6 topological polar surface area; 7 violations of the Lipinski and Veber rules.
Table 6. Predicted pharmacokinetic properties of the most active new quinazolin-4(3H)ones.
Table 6. Predicted pharmacokinetic properties of the most active new quinazolin-4(3H)ones.
Comp.
No.
GIT
Absorption
BBB
Permeability
P-gp SubstrateBioavailability
Score
PAINS
Alert
4aHighNONO0.550
5aHighNONO0.550
5cHighNONO0.550
5dHighNONO0.550
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mohi El-Deen, E.M.; Nossier, E.S.; Karam, E.A. New Quinazolin-4(3H)-one Derivatives Incorporating Hydrazone and Pyrazole Scaffolds as Antimicrobial Agents Targeting DNA Gyraze Enzyme. Sci. Pharm. 2022, 90, 52. https://doi.org/10.3390/scipharm90030052

AMA Style

Mohi El-Deen EM, Nossier ES, Karam EA. New Quinazolin-4(3H)-one Derivatives Incorporating Hydrazone and Pyrazole Scaffolds as Antimicrobial Agents Targeting DNA Gyraze Enzyme. Scientia Pharmaceutica. 2022; 90(3):52. https://doi.org/10.3390/scipharm90030052

Chicago/Turabian Style

Mohi El-Deen, Eman M., Eman S. Nossier, and Eman A. Karam. 2022. "New Quinazolin-4(3H)-one Derivatives Incorporating Hydrazone and Pyrazole Scaffolds as Antimicrobial Agents Targeting DNA Gyraze Enzyme" Scientia Pharmaceutica 90, no. 3: 52. https://doi.org/10.3390/scipharm90030052

Article Metrics

Back to TopTop